1
|
Aroniadou-Anderjaska V, Figueiredo TH, De Araujo Furtado M, Pidoplichko VI, Lumley LA, Braga MFM. Alterations in GABA A receptor-mediated inhibition triggered by status epilepticus and their role in epileptogenesis and increased anxiety. Neurobiol Dis 2024; 200:106633. [PMID: 39117119 DOI: 10.1016/j.nbd.2024.106633] [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: 05/13/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
The triggers of status epilepticus (SE) in non-epileptic patients can vary widely, from idiopathic causes to exposure to chemoconvulsants. Regardless of its etiology, prolonged SE can cause significant brain damage, commonly resulting in the development of epilepsy, which is often accompanied by increased anxiety. GABAA receptor (GABAAR)-mediated inhibition has a central role among the mechanisms underlying brain damage and the ensuing epilepsy and anxiety. During SE, calcium influx primarily via ionotropic glutamate receptors activates signaling cascades which trigger a rapid internalization of synaptic GABAARs; this weakens inhibition, exacerbating seizures and excitotoxicity. GABAergic interneurons are more susceptible to excitotoxic death than principal neurons. During the latent period of epileptogenesis, the aberrant reorganization in synaptic interactions that follow interneuronal loss in injured brain regions, leads to the formation of hyperexcitable, seizurogenic neuronal circuits, along with disturbances in brain oscillatory rhythms. Reduction in the spontaneous, rhythmic "bursts" of IPSCs in basolateral amygdala neurons is likely to play a central role in anxiogenesis. Protecting interneurons during SE is key to preventing both epilepsy and anxiety. Antiglutamatergic treatments, including antagonism of calcium-permeable AMPA receptors, can be expected to control seizures and reduce excitotoxicity not only by directly suppressing hyperexcitation, but also by counteracting the internalization of synaptic GABAARs. Benzodiazepines, as delayed treatment of SE, have low efficacy due to the reduction and dispersion of their targets (the synaptic GABAARs), but also because themselves contribute to further reduction of available GABAARs at the synapse; furthermore, benzodiazepines may be completely ineffective in the immature brain.
Collapse
Affiliation(s)
- Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Marcio De Araujo Furtado
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Volodymyr I Pidoplichko
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Lucille A Lumley
- U.S. Army Medical Research Institute of Chemical Defense, Aberdeen, Proving Ground, MD, USA.
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
2
|
Ghosh A, Quinlan S, Forcelli PA. Anti-seizure medication-induced developmental cell death in neonatal rats is unaltered by history of hypoxia. Epilepsy Res 2024; 201:107318. [PMID: 38430668 PMCID: PMC11018699 DOI: 10.1016/j.eplepsyres.2024.107318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/15/2024] [Accepted: 02/01/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Many anti-seizure medications (ASMs) trigger neuronal cell death when administered during a confined period of early life in rodents. Prototypical ASMs used to treat early-life seizures such as phenobarbital induce this effect, whereas levetiracetam does not. However, most prior studies have examined the effect of ASMs in naïve animals, and the degree to which underlying brain injury interacts with these drugs to modify cell death is poorly studied. Moreover, the degree to which drug-induced neuronal cell death differs as a function of sex is unknown. METHODS We treated postnatal day 7 Sprague Dawley rat pups with vehicle, phenobarbital (75 mg/kg) or levetiracetam (200 mg/kg). Separate groups of pups were pre-exposed to either normoxia or graded global hypoxia. Separate groups of males and females were used. Twenty-four hours after drug treatment, brains were collected and processed for markers of cell death. RESULTS Consistent with prior studies, phenobarbital, but not levetiracetam, increased cell death in cortical regions, basal ganglia, hippocampus, septum, and lateral thalamus. Hypoxia did not modify basal levels of cell death. Females - collapsed across treatment and hypoxia status, displayed a small but significant increase in cell death as compared to males in the cingulate cortex, somatosensory cortex, and the CA1 and CA3 hippocampus; these effects were not modulated by hypoxia or drug treatment. CONCLUSION We found that a history of graded global hypoxia does not alter the neurotoxic profile of phenobarbital. Levetiracetam, which does not induce cell death in normal developing animals, maintained a benign profile on the background of neonatal hypoxia. We found a sex-based difference, as female animals showed elevated levels of cell death across all treatment conditions. Together, these data address several long-standing gaps in our understanding of the neurotoxic profile of antiseizure medications during early postnatal development.
Collapse
Affiliation(s)
- Anjik Ghosh
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA
| | - Sean Quinlan
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA
| | - Patrick A Forcelli
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA; Department of Neuroscience, Georgetown University, Washington, DC, USA; Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA.
| |
Collapse
|
3
|
Goikolea-Vives A, Fernandes C, Thomas MSC, Thornton C, Stolp HB. Sex-Specific Behavioural Deficits in Adulthood following Acute Activation of the GABAA Receptor in the Neonatal Mouse. Dev Neurosci 2024; 46:386-400. [PMID: 38325353 DOI: 10.1159/000536641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/26/2024] [Indexed: 02/09/2024] Open
Abstract
INTRODUCTION Sex differences exist in the prevalence of neurodevelopmental disorders (NDDs). Part of the aetiology of NDDs has been proposed to be alterations in the balance between excitatory and inhibitory neurotransmission, leading to the question of whether males and females respond differently to altered neurotransmitter balance. We investigated whether pharmacological alteration of GABAA signalling in early development results in sex-dependent changes in adult behaviours associated with NDDs. METHODS Male and female C57BL/6J mice received intraperitoneal injections of 0.5 mg/kg muscimol or saline on postnatal days (P) 3-5 and were subjected to behavioural testing, specifically open field, light/dark box, marble-burying, sucralose preference, social interaction, and olfactory habituation/dishabituation tests between P60 and P90. RESULTS Early postnatal administration of muscimol resulted in reduced anxiety in the light/dark box test in both male and female adult mice. Muscimol reduced sucralose preference in males, but not females, whereas female mice showed reduced social behaviours. Regional alterations in cortical thickness were observed in the weeks following GABAA receptor activation, pointing to an evolving structural difference in the brain underlying adult behaviour. CONCLUSIONS We conclude that activation of the GABAA receptor in the first week of life resulted in long-lasting changes in a range of behaviours in adulthood following altered neurodevelopment. Sex of the individual affected the nature and severity of these abnormalities, explaining part of the varied pathophysiology and neurodevelopmental diagnosis that derive from excitatory/inhibitory imbalance.
Collapse
Affiliation(s)
- Ane Goikolea-Vives
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Cathy Fernandes
- SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Michael S C Thomas
- Centre for Brain and Cognitive Development, Birkbeck College, University of London, London, UK
| | - Claire Thornton
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Helen B Stolp
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
4
|
Park SA, Nguyen TTP, Park SJ, Han SK. Naringenin modulates GABA mediated response in a sexdependent manner in substantia gelatinosa neurons of trigeminal subnucleus caudalis in immature mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:73-81. [PMID: 38154966 PMCID: PMC10762483 DOI: 10.4196/kjpp.2024.28.1.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 12/30/2023]
Abstract
The substantia gelatinosa (SG) within the trigeminal subnucleus caudalis (Vc) is recognized as a pivotal site of integrating and modulating afferent fibers carrying orofacial nociceptive information. Although naringenin (4',5,7-thrihydroxyflavanone), a natural bioflavonoid, has been proven to possess various biological effects in the central nervous system (CNS), the activity of naringenin at the orofacial nociceptive site has not been reported yet. In this study, we explored the influence of naringenin on GABA response in SG neurons of Vc using whole-cell patch-clamp technique. The application of GABA in a bath induced two forms of GABA responses: slow and fast. Naringenin enhanced both amplitude and area under curve (AUC) of GABA-mediated responses in 57% (12/21) of tested neurons while decreasing both parameters in 33% (7/21) of neurons. The enhancing or suppressing effect of naringenin on GABA response have been observed, with enhancement occurring when the GABA response was slow, and suppression when it was fast. Furthermore, both the enhancement of slower GABA responses and the suppression of faster GABA responses by naringenin were concentration dependent. Interestingly, the nature of GABA response was also found to be sex-dependent. A majority of SG neurons from juvenile female mice exhibited slower GABA responses, whereas those from juvenile males predominantly displayed faster GABA responses. Taken together, this study indicates that naringenin plays a partial role in modulating orofacial nociception and may hold promise as a therapeutic target for treating orofacial pain, with effects that vary according to sex.
Collapse
Affiliation(s)
- Seon Ah Park
- Department of Oral Physiology, School of Dentistry & Institute of Oral Bioscience, Jeonbuk National University, Jeonju 54896, Korea
| | - Thao Thi Phuong Nguyen
- Faculty of Odonto – Stomatology, Hue University of Medicine and Pharmacy, Hue University, Hue 53000, Vietnam
| | - Soo Joung Park
- Department of Oral Physiology, School of Dentistry & Institute of Oral Bioscience, Jeonbuk National University, Jeonju 54896, Korea
| | - Seong Kyu Han
- Department of Oral Physiology, School of Dentistry & Institute of Oral Bioscience, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
5
|
Campos-Rodriguez C, Palmer D, Forcelli PA. Optogenetic stimulation of the superior colliculus suppresses genetic absence seizures. Brain 2023; 146:4320-4335. [PMID: 37192344 PMCID: PMC11004938 DOI: 10.1093/brain/awad166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 04/18/2023] [Accepted: 05/01/2023] [Indexed: 05/18/2023] Open
Abstract
While anti-seizure medications are effective for many patients, nearly one-third of individuals have seizures that are refractory to pharmacotherapy. Prior studies using evoked preclinical seizure models have shown that pharmacological activation or excitatory optogenetic stimulation of the deep and intermediate layers of the superior colliculus (DLSC) display multi-potent anti-seizure effects. Here we monitored and modulated DLSC activity to suppress spontaneous seizures in the WAG/Rij genetic model of absence epilepsy. Female and male WAG/Rij adult rats were employed as study subjects. For electrophysiology studies, we recorded single unit activity from microwire arrays placed within the DLSC. For optogenetic experiments, animals were injected with virus coding for channelrhodopsin-2 or a control vector, and we compared the efficacy of continuous neuromodulation to that of closed-loop neuromodulation paradigms. For each, we compared three stimulation frequencies on a within-subject basis (5, 20, 100 Hz). For closed-loop stimulation, we detected seizures in real time based on the EEG power within the characteristic frequency band of spike-and-wave discharges (SWDs). We quantified the number and duration of each SWD during each 2 h-observation period. Following completion of the experiment, virus expression and fibre-optic placement was confirmed. We found that single-unit activity within the DLSC decreased seconds prior to SWD onset and increased during and after seizures. Nearly 40% of neurons displayed suppression of firing in response to the start of SWDs. Continuous optogenetic stimulation of the DLSC (at each of the three frequencies) resulted in a significant reduction of SWDs in males and was without effect in females. In contrast, closed-loop neuromodulation was effective in both females and males at all three frequencies. These data demonstrate that activity within the DLSC is suppressed prior to SWD onset, increases at SWD onset, and that excitatory optogenetic stimulation of the DLSC exerts anti-seizure effects against absence seizures. The striking difference between open- and closed-loop neuromodulation approaches underscores the importance of the stimulation paradigm in determining therapeutic effects.
Collapse
Affiliation(s)
| | - Devin Palmer
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007, USA
| | - Patrick A Forcelli
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007, USA
- Department of Neuroscience, Georgetown University, Washington, DC 20007, USA
| |
Collapse
|
6
|
Zhou S, Zhu J, Sun X, Xie L, Zhao Y, Ding S, Wang L, Chen J, Zhu B, Zheng A, Li Y, Zhou C, Shao F. Safety, Pharmacokinetics, and Pharmacodynamics of Midazolam Gel After Rectal Administration in Healthy Chinese Subjects. Clin Drug Investig 2023:10.1007/s40261-023-01276-5. [PMID: 37270744 DOI: 10.1007/s40261-023-01276-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND AND OBJECTIVES Midazolam rectal gel is a novel rectal formulation that may be a promising and potential alternative to oral administration for pediatric sedation. The objective of this study was to evaluate the safety, pharmacokinetics, pharmacodynamics, and absolute bioavailability of midazolam rectal gel in healthy Chinese subjects. METHODS An open-label, single-dose, randomized, two-period, two-treatment, crossover clinical study was conducted in 22 healthy subjects (16 males and six females), each receiving 2.5 mg intravenous midazolam in one period and 5 mg midazolam rectal gel in another period (the dosages here were calculated as active midazolam). Safety, pharmacokinetic, and pharmacodynamic assessments were conducted throughout the study. RESULTS All of the subjects completed both treatment periods. The formulation of rectal gel was well tolerated, with no serious adverse events occurring. After a single rectal dose of 5 mg midazolam rectal gel, it was absorbed rapidly with a median value of time to peak concentration (Tmax) of 1.00 h, and mean values of the peak concentration (Cmax) and area under the concentration-time curve (AUC0-t) of 37.2 ng/mL and 137 h·ng/mL, respectively. The absolute bioavailability of rectal gel was 59.7%. The rectal gel exhibited a relatively delayed onset but a more stable sedative effect and a longer duration when compared with intravenous midazolam. CONCLUSION Midazolam rectal gel may be a feasible alternative with a high level of acceptance in pediatric sedation and enhanced bioavailability compared to an oral formulation. The modeling results may help to disclose out the exposure-response relationship of midazolam rectal gel and support the design of an escalating-doses study and pediatric extrapolation study. CLINICAL TRIAL REGISTRATION The study was registered at http://www.chinadrugtrials.org.cn (No. CTR20192350).
Collapse
Affiliation(s)
- Sufeng Zhou
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jinying Zhu
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaodi Sun
- Department of Anesthesiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Lijun Xie
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yuqing Zhao
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Sijia Ding
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Lu Wang
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Juan Chen
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Bei Zhu
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Aiping Zheng
- Department of Pharmaceutics, Institute of Pharmacology and Toxicology of Academy of Military Medical Sciences, Beijing, 100850, China
| | - Yajuan Li
- Xinjiang Tefeng Pharmaceutical Company, Ltd., Urumqi, 830054, China
| | - Chen Zhou
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| | - Feng Shao
- Phase I Clinical Trial Unit, The First Affiliated Hospital with Nanjing Medical University, #300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
7
|
Sharma D, Tripathi M, Doddamani R, Sharma MC, Lalwani S, Sarat Chandra P, Banerjee Dixit A, Banerjee J. Correlation of age at seizure onset with GABA A receptor subunit and chloride Co-transporter configuration in Focal cortical dysplasia (FCD). Neurosci Lett 2023; 796:137065. [PMID: 36638954 DOI: 10.1016/j.neulet.2023.137065] [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: 09/14/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Focal cortical dysplasia (FCD) represents a group of malformations of cortical development, which are speculated to be related to early developmental defects in the cerebral cortex. According to dysmature cerebral development hypothesis of FCD altered GABAA receptor function is known to contribute to abnormal neuronal network. Here, we studied the possible association between age at seizure onset in FCD with the subunit configuration of GABAA receptors in resected brain specimens obtained from patients with FCD. We observed a significantly higher ratio of α4/α1 subunit-containing GABAA receptors in patients with early onset (EO) FCD as compared to those with late onset (LO) FCD as is seen during the course of development where α4-containing GABAA receptors expression is high as compared to α1-containing GABAA receptors expression. Likewise, the influx to efflux chloride co-transporter expression of NKCC1/KCC2 was also increased in patients with EO FCD as seen during brain development. In addition, we observed that the ratio of GABA/Glutamate neurotransmitters was lower in patients with EO FCD as compared to that in patients with LO FCD. Our findings suggest altered configuration of GABAA receptors in FCD which could be contributing to aberrant depolarizing GABAergic activity. In particular, we observed a correlation of age at seizure onset in FCD with subunit configuration of GABAA receptors, levels of NKCC1/KCC2 and the ratio of GABA/Glutamate neurotransmitters such that the patients with EO FCD exhibited a more critically modulated GABAergic network.
Collapse
Affiliation(s)
- Devina Sharma
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Ramesh Doddamani
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - M C Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjeev Lalwani
- Department of Forensic Medicine and Toxicology, All India Institute of Medical Sciences, New Delhi, India
| | - P Sarat Chandra
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Aparna Banerjee Dixit
- Dr. B.R Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Jyotirmoy Banerjee
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
8
|
Ruiter M, Herstel LJ, Wierenga CJ. Reduction of Dendritic Inhibition in CA1 Pyramidal Neurons in Amyloidosis Models of Early Alzheimer's Disease. J Alzheimers Dis 2021; 78:951-964. [PMID: 33074225 PMCID: PMC7739974 DOI: 10.3233/jad-200527] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: In an early stage of Alzheimer’s disease (AD), before the formation of amyloid plaques, neuronal network hyperactivity has been reported in both patients and animal models. This suggests an underlying disturbance of the balance between excitation and inhibition. Several studies have highlighted the role of somatic inhibition in early AD, while less is known about dendritic inhibition. Objective: In this study we investigated how inhibitory synaptic currents are affected by elevated Aβ levels. Methods: We performed whole-cell patch clamp recordings of CA1 pyramidal neurons in organotypic hippocampal slice cultures after treatment with Aβ-oligomers and in hippocampal brain slices from AppNL-F-G mice (APP-KI). Results: We found a reduction of spontaneous inhibitory postsynaptic currents (sIPSCs) in CA1 pyramidal neurons in organotypic slices after 24 h Aβ treatment. sIPSCs with slow rise times were reduced, suggesting a specific loss of dendritic inhibitory inputs. As miniature IPSCs and synaptic density were unaffected, these results suggest a decrease in activity-dependent transmission after Aβ treatment. We observed a similar, although weaker, reduction in sIPSCs in CA1 pyramidal neurons from APP-KI mice compared to control. When separated by sex, the strongest reduction in sIPSC frequency was found in slices from male APP-KI mice. Consistent with hyperexcitability in pyramidal cells, dendritically targeting interneurons received slightly more excitatory input. GABAergic action potentials had faster kinetics in APP-KI slices. Conclusion: Our results show that Aβ affects dendritic inhibition via impaired action potential driven release, possibly due to altered kinetics of GABAergic action potentials. Reduced dendritic inhibition may contribute to neuronal hyperactivity in early AD.
Collapse
Affiliation(s)
- Marvin Ruiter
- Department of Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Lotte J Herstel
- Department of Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Corette J Wierenga
- Department of Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
9
|
González EA, Calsbeek JJ, Tsai YH, Tang MY, Andrew P, Vu J, Berg EL, Saito NH, Harvey DJ, Supasai S, Gurkoff GG, Silverman JL, Lein PJ. Sex-specific acute and chronic neurotoxicity of acute diisopropylfluorophosphate (DFP)-intoxication in juvenile Sprague-Dawley rats. Curr Res Toxicol 2021; 2:341-356. [PMID: 34622217 PMCID: PMC8484742 DOI: 10.1016/j.crtox.2021.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Preclinical efforts to improve medical countermeasures against organophosphate (OP) chemical threat agents have largely focused on adult male models. However, age and sex have been shown to influence the neurotoxicity of repeated low-level OP exposure. Therefore, to determine the influence of sex and age on outcomes associated with acute OP intoxication, postnatal day 28 Sprague-Dawley male and female rats were exposed to the OP diisopropylfluorophosphate (DFP; 3.4 mg/kg, s.c.) or an equal volume of vehicle (∼80 µL saline, s.c.) followed by atropine sulfate (0.1 mg/kg, i.m.) and pralidoxime (2-PAM; 25 mg/kg, i.m.). Seizure activity was assessed during the first 4 h post-exposure using behavioral criteria and electroencephalographic (EEG) recordings. At 1 d post-exposure, acetylcholinesterase (AChE) activity was measured in cortical tissue, and at 1, 7, and 28 d post-exposure, brains were collected for neuropathologic analyses. At 1 month post-DFP, animals were analyzed for motor ability, learning and memory, and hippocampal neurogenesis. Acute DFP intoxication triggered more severe seizure behavior in males than females, which was supported by EEG recordings. DFP caused significant neurodegeneration and persistent microglial activation in numerous brain regions of both sexes, but astrogliosis occurred earlier and was more severe in males compared to females. DFP males and females exhibited pronounced memory deficits relative to sex-matched controls. In contrast, acute DFP intoxication altered hippocampal neurogenesis in males, but not females. These findings demonstrate that acute DFP intoxication triggers seizures in juvenile rats of both sexes, but the seizure severity varies by sex. Some, but not all, chronic neurotoxic outcomes also varied by sex. The spatiotemporal patterns of neurological damage suggest that microglial activation may be a more important factor than astrogliosis or altered neurogenesis in the pathogenesis of cognitive deficits in juvenile rats acutely intoxicated with OPs.
Collapse
Key Words
- 2-PAM, pralidoxime
- AChE, acetylcholinesterase
- AS, atropine-sulfate
- BChE, butyrylcholinesterase
- CT, computed tomography
- ChE, cholinesterase
- Cognitive deficits
- DFP, diisopropylfluorophosphate
- EEG, electroencephalogram
- FJC, Fluoro-Jade C
- Neurodegeneration
- Neurogenesis
- Neuroinflammation
- OP, organophosphate
- PBS, phosphate-buffered saline
- ROI, region of interest
- SE, status epilepticus
- Seizures
- Sex differences
- T2w, T2-weighted
- VEH, vehicle
- i.m., intramuscular
- i.p., intraperitoneal
- s.c., subcutaneous
Collapse
Affiliation(s)
- Eduardo A. González
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Jonas J. Calsbeek
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Yi-Hua Tsai
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Mei-Yun Tang
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Peter Andrew
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Joan Vu
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Elizabeth L. Berg
- Department of Psychiatry, University of California, Davis, School of Medicine, 2230, Stockton Boulevard, Sacramento, CA 95817, USA
| | - Naomi H. Saito
- Department of Public Health Sciences, University of California, Davis, One Shields Avenue, School of Medicine, Davis, CA 95616, USA
| | - Danielle J. Harvey
- Department of Public Health Sciences, University of California, Davis, One Shields Avenue, School of Medicine, Davis, CA 95616, USA
| | - Suangsuda Supasai
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| | - Gene G. Gurkoff
- Department of Neurological Surgery, University of California, Davis, School of Medicine, 4860 Y Street, Sacramento, CA 95817, USA
- Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA 95618, USA
| | - Jill L. Silverman
- Department of Psychiatry, University of California, Davis, School of Medicine, 2230, Stockton Boulevard, Sacramento, CA 95817, USA
- MIND Institute, University of California, Davis, 2825 50th Street, Sacramento, CA 95817, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
- MIND Institute, University of California, Davis, 2825 50th Street, Sacramento, CA 95817, USA
| |
Collapse
|
10
|
Rahman MA, Keck TM, Poe MM, Sharmin D, Cook JM, Fischer BD. Synergistic antihyperalgesic and antinociceptive effects of morphine and methyl 8-ethynyl-6-(pyridin-2-yl)-4H-benzo[f]imidazo[1,5-a][1,4]diazepine-3-carboxylate (MP-III-024): a positive allosteric modulator at α2GABA A and α3GABA A receptors. Psychopharmacology (Berl) 2021; 238:1585-1592. [PMID: 33585961 PMCID: PMC8141038 DOI: 10.1007/s00213-021-05791-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/04/2021] [Indexed: 11/24/2022]
Abstract
RATIONALE Opioid and GABAA receptors are both located in central nociceptive pathways, and compounds that activate these receptors have pain-relieving properties. To date, the interactive effects of concurrent administration of these compounds in preclinical models of pain-like behaviors have not been assessed. OBJECTIVE The purpose of this study was to examine the interactive effects of the μ-opioid agonist morphine and the α2GABAA and α3GABAA receptor positive allosteric modulator methyl 8-ethynyl-6-(pyridin-2-yl)-4H-benzo[f]imidazo[1,5-a][1,4]diazepine-3-carboxylate (MP-III-024) in preclinical models of mechanical hyperalgesia and thermal nociception. METHODS The antihyperalgesic and antinociceptive effects of morphine and MP-III-024 administered alone were assessed initially, followed by fixed-ratio mixtures of MP-III-024/morphine combinations. Drug interaction data were analyzed using isobolographic and dose-addition analyses. All studies were conducted in male CD-1 mice. RESULTS In the assay of mechanical hyperalgesia, each compound produced dose-dependent antihyperalgesic effects, whereas only morphine was effective on thermal nociception. Fixed-ratio mixtures of MP-III-024/morphine were also dose-dependently effective in both procedures. These drug combination studies revealed that morphine and MP-III-024 produced supra-additive (synergistic) effects in both assays, depending on their relative proportions. CONCLUSIONS These results demonstrate an interaction between α2GABAA and α3GABAA receptor- and μ-opioid receptor-mediated signals and suggest that combination therapy may be useful for the treatment of pain-related disorders.
Collapse
Affiliation(s)
- Mohammad A. Rahman
- Rowan University, Department of Chemistry & Biochemistry, Department of Molecular & Cellular Biosciences, Glassboro, NJ 08028, USA
| | - Thomas M. Keck
- Rowan University, Department of Chemistry & Biochemistry, Department of Molecular & Cellular Biosciences, Glassboro, NJ 08028, USA
| | - Michael M. Poe
- University of Wisconsin-Milwaukee, Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, Milwaukee, Wisconsin 53201, USA
| | - Dishary Sharmin
- University of Wisconsin-Milwaukee, Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, Milwaukee, Wisconsin 53201, USA
| | - James M. Cook
- University of Wisconsin-Milwaukee, Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, Milwaukee, Wisconsin 53201, USA
| | - Bradford D. Fischer
- Cooper Medical School of Rowan University, Department of Biomedical Sciences Camden, NJ 08103, USA,Corresponding Author:; Phone: (856) 361-2869
| |
Collapse
|
11
|
Christian CA, Reddy DS, Maguire J, Forcelli PA. Sex Differences in the Epilepsies and Associated Comorbidities: Implications for Use and Development of Pharmacotherapies. Pharmacol Rev 2020; 72:767-800. [PMID: 32817274 PMCID: PMC7495340 DOI: 10.1124/pr.119.017392] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The epilepsies are common neurologic disorders characterized by spontaneous recurrent seizures. Boys, girls, men, and women of all ages are affected by epilepsy and, in many cases, by associated comorbidities as well. The primary courses of treatment are pharmacological, dietary, and/or surgical, depending on several factors, including the areas of the brain affected and the severity of the epilepsy. There is a growing appreciation that sex differences in underlying brain function and in the neurobiology of epilepsy are important factors that should be accounted for in the design and development of new therapies. In this review, we discuss the current knowledge on sex differences in epilepsy and associated comorbidities, with emphasis on those aspects most informative for the development of new pharmacotherapies. Particular focus is placed on sex differences in the prevalence and presentation of various focal and generalized epilepsies; psychiatric, cognitive, and physiologic comorbidities; catamenial epilepsy in women; sex differences in brain development; the neural actions of sex and stress hormones and their metabolites; and cellular mechanisms, including brain-derived neurotrophic factor signaling and neuronal-glial interactions. Further attention placed on potential sex differences in epilepsies, comorbidities, and drug effects will enhance therapeutic options and efficacy for all patients with epilepsy. SIGNIFICANCE STATEMENT: Epilepsy is a common neurological disorder that often presents together with various comorbidities. The features of epilepsy and seizure activity as well as comorbid afflictions can vary between men and women. In this review, we discuss sex differences in types of epilepsies, associated comorbidities, pathophysiological mechanisms, and antiepileptic drug efficacy in both clinical patient populations and preclinical animal models.
Collapse
Affiliation(s)
- Catherine A Christian
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Doodipala Samba Reddy
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Jamie Maguire
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| | - Patrick A Forcelli
- Department of Molecular and Integrative Physiology, Neuroscience Program, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois (C.A.C.); Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas (D.S.R.); Neuroscience Department, Tufts University School of Medicine, Boston, Massachusetts (J.M.); and Departments of Pharmacology and Physiology and Neuroscience, Georgetown University, Washington, D.C. (P.A.F.)
| |
Collapse
|
12
|
Niibori Y, Lee SJ, Minassian BA, Hampson DR. Sexually Divergent Mortality and Partial Phenotypic Rescue After Gene Therapy in a Mouse Model of Dravet Syndrome. Hum Gene Ther 2020; 31:339-351. [PMID: 31830809 PMCID: PMC7087406 DOI: 10.1089/hum.2019.225] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Dravet syndrome (DS) is a neurodevelopmental genetic disorder caused by mutations in the SCN1A gene encoding the α subunit of the NaV1.1 voltage-gated sodium channel that controls neuronal action potential firing. The high density of this mutated channel in GABAergic interneurons results in impaired inhibitory neurotransmission and subsequent excessive activation of excitatory neurons. The syndrome is associated with severe childhood epilepsy, autistic behaviors, and sudden unexpected death in epilepsy. Here, we compared the rescue effects of an adeno-associated viral (AAV) vector coding for the multifunctional β1 sodium channel auxiliary subunit (AAV-NaVβ1) with a control vector lacking a transgene. We hypothesized that overexpression of NaVβ1 would facilitate the function of residual voltage-gated channels and improve the DS phenotype in the Scn1a+/− mouse model of DS. AAV-NaVβ1 was injected into the cerebral spinal fluid of neonatal Scn1a+/− mice. In untreated control Scn1a+/− mice, females showed a higher degree of mortality than males. Compared with Scn1a+/− control mice, AAV-NaVβ1-treated Scn1a+/− mice displayed increased survival, an outcome that was more pronounced in females than males. In contrast, behavioral analysis revealed that male, but not female, Scn1a+/− mice displayed motor hyperactivity, and abnormal performance on tests of fear and anxiety and learning and memory. Male Scn1a+/− mice treated with AAV-NaVβ1 showed reduced spontaneous seizures and normalization of motor activity and performance on the elevated plus maze test. These findings demonstrate sex differences in mortality in untreated Scn1a+/− mice, an effect that may be related to a lower level of intrinsic inhibitory tone in female mice, and a normalization of aberrant behaviors in males after central nervous system administration of AAV-NaVβ1. The therapeutic efficacy of AAV-NaVβ1 in a mouse model of DS suggests a potential new long-lasting biological therapeutic avenue for the treatment of this catastrophic epilepsy.
Collapse
Affiliation(s)
- Yosuke Niibori
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Shiron J Lee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Berge A Minassian
- Department of Pediatrics, University of Texas Southwest Medical Center, Dallas, Texas
| | - David R Hampson
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada.,Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
13
|
El Amki M, Binder N, Steffen R, Schneider H, Luft AR, Weller M, Imthurn B, Merki-Feld GS, Wegener S. Contraceptive drugs mitigate experimental stroke-induced brain injury. Cardiovasc Res 2018; 115:637-646. [DOI: 10.1093/cvr/cvy248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
AbstractAimsEffective stroke treatments beyond reperfusion remain scant. The natural steroid hormone progesterone has shown protective effects in experimental models of brain injury and cardiovascular disease. However, unfavourable bioavailability limits its clinical use. Desogestrel and drospirenone are new generation progestins with progesterone-like properties, developed as oral contraceptives with excellent bioavailability and safety profile. We investigated the neuroprotective properties of these progestins in vivo using transient middle cerebral artery occlusion (MCAO) and in vitro using an oxygen-glucose deprivation and reoxygenation (OGD/R) model in primary neuronal cells.Methods and resultsMCAO was induced in female, female ovariectomized (modelling postmenopausal females) and male mice. Treatment with the progestins resulted in less severe strokes after MCAO and less neuronal death in OGD/R. Desogestrel and drospirenone induced higher expression levels of GABAAR α4 and delta subunits within the brain, suggesting changes in GABAAR configuration favouring tonic inhibition as potential mechanism of action. Treatment with the GABAAR blocker picrotoxin abolished the protection afforded by the progestins in vivo and in vitro.ConclusionFor the first time, here, we delineate a potential role of desogestrel and drospirenone, both clinically approved and safe drugs in mitigating the consequences of stroke. Contraception with desogestrel and drospirenone in progestin-only preparations may be particularly beneficial for women at risk of stroke.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Nadine Binder
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Riccardo Steffen
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Hannah Schneider
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Andreas R Luft
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Bruno Imthurn
- Department of Reproductive Endocrinology, University Hospital Zurich, Switzerland
| | | | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| |
Collapse
|
14
|
Moshé SL. The 2017 Sachs Lecture: Kindling Knowledge in Epilepsy. Pediatr Neurol 2018; 85:5-12. [PMID: 29958806 DOI: 10.1016/j.pediatrneurol.2018.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Solomon L Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx, New York; Dominick P. Purpura Department of Neuroscience, Montefiore/Einstein Epilepsy Center, Albert Einstein College of Medicine, Bronx, New York; Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
15
|
Samba Reddy D. Sex differences in the anticonvulsant activity of neurosteroids. J Neurosci Res 2017; 95:661-670. [PMID: 27870400 DOI: 10.1002/jnr.23853] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/21/2016] [Accepted: 07/06/2016] [Indexed: 12/11/2022]
Abstract
Epilepsy is one of the leading causes of chronic neurological morbidity worldwide. Acquired epilepsy may result from a number of conditions, such as brain injury, anoxia, tumors, stroke, neurotoxicity, and prolonged seizures. Sex differences have been observed in many seizure types; however, some sex-specific seizure disorders are much more prevalent in women. Despite some inconsistencies, substantial data indicates that sensitivity to seizure stimuli differs between the sexes. Men generally exhibit greater seizure susceptibility than women, whereas many women with epilepsy experience a cyclical occurrence of seizures that tends to center around the menstrual period, which has been termed catamenial epilepsy. Some epilepsy syndromes show gender differences with female predominance or male predominance. Steroid hormones, endogenous neurosteroids, and sexually dimorphic neural networks appear to play a key role in sex differences in seizure susceptibility. Neurosteroids, such as allopregnanolone, reflect sex differences in their anticonvulsant activity. This Review provides a brief overview of the evidence for sex differences in epilepsy and how sex differences influence the use of neurosteroids in epilepsy and epileptogenesis. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Sciences Center, College of Medicine, Bryan, Texas
| |
Collapse
|
16
|
Changes in GABAergic markers accompany degradation of neuronal function in the primary visual cortex of senescent rats. Sci Rep 2017; 7:14897. [PMID: 29097694 PMCID: PMC5668371 DOI: 10.1038/s41598-017-15006-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/19/2017] [Indexed: 11/14/2022] Open
Abstract
Numerous studies have reported age-dependent degradation of neuronal function in the visual cortex and have attributed this functional decline to weakened intracortical inhibition, especially GABAergic inhibition. However, whether this type of functional decline is linked to compromised GABAergic inhibition has not been fully confirmed. Here, we compared the neuronal response properties and markers of GABAergic inhibition in the primary visual cortex (V1) of young adult and senescent rats. Compared with those of young adult rats, old rats’ V1 neurons exhibited significantly increased visually evoked responses and spontaneous activity, a decreased signal-to-noise ratio and reduced response selectivity for the stimulus orientation and motion direction. Additionally, the ratio of GABA-positive neurons to total cortical neurons in old rats was significantly decreased compared with that in young rats. Expression of the key GABA-synthesizing enzyme GAD67 was significantly lower in old rats than in young rats, although GAD65 expression showed a marginal difference between the two age groups. Further, expression of an important GABAA receptor subunit, GABAAR α1, was significantly attenuated in old rats relative to young ones. These results demonstrate that ageing may result in decreased GABAergic inhibition in the visual cortex and that this decrease in GABAergic inhibition accompanies neuronal function degradation.
Collapse
|
17
|
Ferreri F, Guerra A, Vollero L, Ponzo D, Maatta S, Mervaala E, Iannello G, Di Lazzaro V. Age-related changes of cortical excitability and connectivity in healthy humans: non-invasive evaluation of sensorimotor network by means of TMS-EEG. Neuroscience 2017. [DOI: 10.1016/j.neuroscience.2017.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Florinda Ferreri
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy; Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, 70029 KYS, Kuopio, Finland.
| | - Andrea Guerra
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Luca Vollero
- Department of Computer Science and Computer Engineering, University Campus Bio-Medico, via Álvaro del Portillo 200, 00128 Rome, Italy
| | - David Ponzo
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy
| | - Sara Maatta
- Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, 70029 KYS, Kuopio, Finland
| | - Esa Mervaala
- Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, 70029 KYS, Kuopio, Finland
| | - Giulio Iannello
- Department of Computer Science and Computer Engineering, University Campus Bio-Medico, via Álvaro del Portillo 200, 00128 Rome, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, via Álvaro del Portillo 21, 00128 Rome, Italy
| |
Collapse
|
18
|
Developmental pharmacology of benzodiazepines under normal and pathological conditions. Epileptic Disord 2016; 16 Spec No 1:S59-68. [PMID: 25335485 DOI: 10.1684/epd.2014.0690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Benzodiazepines are allosteric agonists of GABAA receptors (GABAAR), pentameric ligand-gated Cl(-) channels, which serve both an important neurodevelopmental role but are also the principal inhibitory system in the brain. However, their subunit composition, channel properties, and function, as well as their region-specific expression patterns, change through development. These processes have been extensively studied in rodents and to some extent confirmed in higher species. Specifically, GABAARs acquire faster kinetics with age and their pharmacology changes rendering them more sensitive to drugs that have higher affinity for α1 subunit-containing GABAARs, such as benzodiazepines, but also, their inhibitory function becomes more potent as they shift from having depolarising to hyperpolarising responses due to a shift in Cl(-) gradient and cation chloride cotransporter expression. Concerns have been raised about possible pro-apoptotic and paradoxical effects of benzodiazepines in the neonatal normal rat brain, although it is unclear, as yet, whether this extends to brains exposed to seizures. Growing evidence indicates that the pharmacology and physiology of GABAARs may be altered in the brain of rats or humans with seizures or epilepsy, or different aetiologies that predispose to epilepsy. These changes follow different paths, depending on sex, age, region, cell type, aetiology, or time-point specific factors. Identification of dynamic biomarkers that could enable these changes in vivo to be monitored would greatly facilitate the selection of more effective agonists with fewer side effects.
Collapse
|
19
|
Nishimura Y, Okabe S, Sasagawa S, Murakami S, Ashikawa Y, Yuge M, Kawaguchi K, Kawase R, Tanaka T. Pharmacological profiling of zebrafish behavior using chemical and genetic classification of sleep-wake modifiers. Front Pharmacol 2015; 6:257. [PMID: 26578964 PMCID: PMC4630575 DOI: 10.3389/fphar.2015.00257] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/19/2015] [Indexed: 01/05/2023] Open
Abstract
Sleep-wake states are impaired in various neurological disorders. Impairment of sleep-wake states can be an early condition that exacerbates these disorders. Therefore, treating sleep-wake dysfunction may prevent or slow the development of these diseases. Although many gene products are likely to be involved in the sleep-wake disturbance, hypnotics and psychostimulants clinically used are limited in terms of their mode of action and are not without side effects. Therefore, there is a growing demand for developing new hypnotics and psychostimulants with high efficacy and few side effects. Toward this end, animal models are indispensable for use in genetic and chemical screens to identify sleep-wake modifiers. As a proof-of-concept study, we performed behavioral profiling of zebrafish treated with chemical and genetic sleep-wake modifiers. We were able to demonstrate that behavioral profiling of zebrafish treated with hypnotics or psychostimulants from 9 to 10 days post-fertilization was sufficient to identify drugs with specific modes of action. We were also able to identify behavioral endpoints distinguishing GABA-A modulators and hypocretin (hcrt) receptor antagonists and between sympathomimetic and non-sympathomimetic psychostimulants. This behavioral profiling can serve to identify genes related to sleep-wake disturbance associated with various neuropsychiatric diseases and novel therapeutic compounds for insomnia and excessive daytime sleep with fewer adverse side effects.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan ; Mie University Medical Zebrafish Research Center Tsu, Japan ; Department of Systems Pharmacology, Mie University Graduate School of Medicine Tsu, Japan ; Department of Omics Medicine, Mie University Industrial Technology Innovation Institute Tsu, Japan ; Department of Bioinformatics, Mie University Life Science Research Center Tsu, Japan
| | - Shiko Okabe
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan
| | - Shota Sasagawa
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan
| | - Soichiro Murakami
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan
| | - Yoshifumi Ashikawa
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan
| | - Mizuki Yuge
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan
| | - Koki Kawaguchi
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan
| | - Reiko Kawase
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan
| | - Toshio Tanaka
- Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Mie University Graduate School of Medicine Tsu, Japan ; Mie University Medical Zebrafish Research Center Tsu, Japan ; Department of Systems Pharmacology, Mie University Graduate School of Medicine Tsu, Japan ; Department of Omics Medicine, Mie University Industrial Technology Innovation Institute Tsu, Japan ; Department of Bioinformatics, Mie University Life Science Research Center Tsu, Japan
| |
Collapse
|
20
|
Age- and sex-related characteristics of tonic GABA currents in the rat substantia nigra pars reticulata. Neurochem Res 2015; 40:747-57. [PMID: 25645446 DOI: 10.1007/s11064-015-1523-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 12/19/2022]
Abstract
Previous studies have shown that the pharmacologic effects of GABAergic drugs and the postsynaptic phasic GABAAergic inhibitory responses in the anterior part of the rat substantia nigra pars reticulata (SNRA) are age- and sex-specific. Here, we investigate whether there are age- and sex-related differences in the expression of the δ GABAA receptor (GABAAR) subunit and GABAAR mediated tonic currents. We have used δ-specific immunochemistry and whole cell patch clamp to study GABAAR mediated tonic currents in the SNRA of male and female postnatal day (PN) PN5-9, PN11-16, and PN25-32 rats. We observed age-related decline, but no sex-specific changes, in bicuculline (BIM) sensitive GABAAR tonic current density, which correlated with the decline in δ subunit in the SNRA between PN15 and 30. Furthermore, we show that the GABAAR tonic currents can be modified by muscimol (GABAAR agonist; partial GABACR agonist), THIP (4,5,6,7-tetrahydroisoxazolo (5,4-c)pyridin-3-ol: α4β3δ GABAARs agonist and GABACR antagonist), and zolpidem (α1-subunit selective GABAAR agonist) in age- and sex-dependent manner specific for each drug. We propose that the emergence of the GABAAR-sensitive anticonvulsant effects of the rat SNRA during development may depend upon the developmental decline in tonic GABAergic inhibition of the activity of rat SNRA neurons, although other sex-specific factors are also involved.
Collapse
|
21
|
Akman O, Moshé SL, Galanopoulou AS. Sex-specific consequences of early life seizures. Neurobiol Dis 2014; 72 Pt B:153-66. [PMID: 24874547 DOI: 10.1016/j.nbd.2014.05.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/07/2014] [Accepted: 05/17/2014] [Indexed: 12/29/2022] Open
Abstract
Seizures are very common in the early periods of life and are often associated with poor neurologic outcome in humans. Animal studies have provided evidence that early life seizures may disrupt neuronal differentiation and connectivity, signaling pathways, and the function of various neuronal networks. There is growing experimental evidence that many signaling pathways, like GABAA receptor signaling, the cellular physiology and differentiation, or the functional maturation of certain brain regions, including those involved in seizure control, mature differently in males and females. However, most experimental studies of early life seizures have not directly investigated the importance of sex on the consequences of early life seizures. The sexual dimorphism of the developing brain raises the question that early seizures could have distinct effects in immature females and males that are subjected to seizures. We will first discuss the evidence for sex-specific features of the developing brain that could be involved in modifying the susceptibility and consequences of early life seizures. We will then review how sex-related biological factors could modify the age-specific consequences of induced seizures in the immature animals. These include signaling pathways (e.g., GABAA receptors), steroid hormones, growth factors. Overall, there are very few studies that have specifically addressed seizure outcomes in developing animals as a function of sex. The available literature indicates that a variety of outcomes (histopathological, behavioral, molecular, epileptogenesis) may be affected in a sex-, age-, region-specific manner after seizures during development. Obtaining a better understanding for the gender-related mechanisms underlying epileptogenesis and seizure comorbidities will be necessary to develop better gender and age appropriate therapies.
Collapse
Affiliation(s)
- Ozlem Akman
- Department of Physiology, Faculty of Medicine, Istanbul Bilim University, 34394 Istanbul, Turkey.
| | - Solomon L Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Montefiore Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA; Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Montefiore Epilepsy Management Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA
| |
Collapse
|
22
|
Giorgi FS, Galanopoulou AS, Moshé SL. Sex dimorphism in seizure-controlling networks. Neurobiol Dis 2014; 72 Pt B:144-52. [PMID: 24851800 DOI: 10.1016/j.nbd.2014.05.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 11/27/2022] Open
Abstract
Males and females show a different predisposition to certain types of seizures in clinical studies. Animal studies have provided growing evidence for sexual dimorphism of certain brain regions, including those that control seizures. Seizures are modulated by networks involving subcortical structures, including thalamus, reticular formation nuclei, and structures belonging to the basal ganglia. In animal models, the substantia nigra pars reticulata (SNR) is the best studied of these areas, given its relevant role in the expression and control of seizures throughout development in the rat. Studies with bilateral infusions of the GABA(A) receptor agonist muscimol have identified distinct roles of the anterior or posterior rat SNR in flurothyl seizure control, that follow sex-specific maturational patterns during development. These studies indicate that (a) the regional functional compartmentalization of the SNR appears only after the third week of life, (b) only the male SNR exhibits muscimol-sensitive proconvulsant effects which, in older animals, is confined to the posterior SNR, and (c) the expression of the muscimol-sensitive anticonvulsant effects become apparent earlier in females than in males. The first three postnatal days are crucial in determining the expression of the muscimol-sensitive proconvulsant effects of the immature male SNR, depending on the gonadal hormone setting. Activation of the androgen receptors during this early period seems to be important for the formation of this proconvulsant SNR region. We describe molecular/anatomical candidates underlying these age- and sex-related differences, as derived from in vitro and in vivo experiments, as well as by [(14)C]2-deoxyglucose autoradiography. These involve sex-specific patterns in the developmental changes in the structure or physiology or GABA(A) receptors or of other subcortical structures (e.g., locus coeruleus, hippocampus) that may affect the function of seizure-controlling networks.
Collapse
Affiliation(s)
- Fillippo Sean Giorgi
- Department of Clinical and Experimental Medicine, Section of Neurology, University of Pisa-Pisa University Hospital, I56126 Pisa, Italy.
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Montefiore Epilepsy Management Center, and Dominick P. Purpura Department of Neuroscience, Bronx, NY, 10461, USA
| | - Solomon L Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Montefiore Epilepsy Management Center, and Dominick P. Purpura Department of Neuroscience, Bronx, NY, 10461, USA; Department of Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, 10461, USA
| |
Collapse
|
23
|
Galanopoulou AS, Moshé SL. Does epilepsy cause a reversion to immature function? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 813:195-209. [PMID: 25012378 DOI: 10.1007/978-94-017-8914-1_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Seizures have variable effects on brain. Numerous studies have examined the consequences of seizures, in light of the way that these may alter the susceptibility of the brain to seizures, promote epileptogenesis, or functionally alter brain leading to seizure-related comorbidities. In many -but not all- situations, seizures shift brain function towards a more immature state, promoting the birth of newborn neurons, altering the dendritic structure and neuronal connectivity, or changing neurotransmitter signaling towards more immature patterns. These effects depend upon many factors, including the seizure type, age of seizure occurrence, sex, and brain region studied. Here we discuss some of these findings proposing that these seizure-induced immature features do not simply represent rejuvenation of the brain but rather a de-synchronization of the homeostatic mechanisms that were in place to maintain normal physiology, which may contribute to epileptogenesis or the cognitive comorbidities.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, The Laboratory of Developmental Epilepsy, Comprehensive Einstein/Montefiore Epilepsy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Kennedy Center Rm 306, Bronx, NY, 10461, USA,
| | | |
Collapse
|
24
|
Huang X, McMahon J, Yang J, Shin D, Huang Y. Rapamycin down-regulates KCC2 expression and increases seizure susceptibility to convulsants in immature rats. Neuroscience 2012; 219:33-47. [PMID: 22613737 PMCID: PMC3402618 DOI: 10.1016/j.neuroscience.2012.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 05/02/2012] [Accepted: 05/03/2012] [Indexed: 11/30/2022]
Abstract
Seizure susceptibility to neurological insults, including chemical convulsants, is age-dependent and most likely reflective of overall differences in brain excitability. The molecular and cellular mechanisms underlying development-dependent seizure susceptibility remain to be fully understood. Because the mammalian target of rapamycin (mTOR) pathway regulates neurite outgrowth, synaptic plasticity and cell survival, thereby influencing brain development, we tested if exposure of the immature brain to the mTOR inhibitor rapamycin changes seizure susceptibility to neurological insults. We found that inhibition of mTOR by rapamycin in immature rats (3-4 weeks old) increases the severity of seizures induced by pilocarpine, including lengthening the total seizure duration and reducing the latency to the onset of seizures. Rapamycin also reduces the minimal dose of pentylenetetrazol (PTZ) necessary to induce clonic seizures. However, in mature rats, rapamycin does not significantly change the seizure sensitivity to pilocarpine and PTZ. Likewise, kainate sensitivity was not significantly affected by rapamycin treatment in either mature or immature rats. Additionally, rapamycin treatment down-regulates the expression of potassium-chloride cotransporter 2 (KCC2) in the thalamus and to a lesser degree in the hippocampus. Pharmacological inhibition of thalamic mTOR or KCC2 increases susceptibility to pilocarpine-induced seizure in immature rats. Thus, our study suggests a role for the mTOR pathway in age-dependent seizure susceptibility.
Collapse
Affiliation(s)
- Xiaoxing Huang
- Center for Neuropharmacology and Neuroscience, Albany Medical College, NY, USA
| | - John McMahon
- Center for Neuropharmacology and Neuroscience, Albany Medical College, NY, USA
| | - Jun Yang
- Center for Neuropharmacology and Neuroscience, Albany Medical College, NY, USA
| | - Damian Shin
- Center for Neuropharmacology and Neuroscience, Albany Medical College, NY, USA
| | - Yunfei Huang
- Center for Neuropharmacology and Neuroscience, Albany Medical College, NY, USA
| |
Collapse
|
25
|
Martin N, Lafortune M, Godbout J, Barakat M, Robillard R, Poirier G, Bastien C, Carrier J. Topography of age-related changes in sleep spindles. Neurobiol Aging 2012; 34:468-76. [PMID: 22809452 DOI: 10.1016/j.neurobiolaging.2012.05.020] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 03/26/2012] [Accepted: 05/27/2012] [Indexed: 10/28/2022]
Abstract
Aging induces multiple changes to sleep spindles, which may hinder their alleged functional role in memory and sleep protection mechanisms. Brain aging in specific cortical regions could affect the neural networks underlying spindle generation, yet the topography of these age-related changes is currently unknown. In the present study, we analyzed spindle characteristics in 114 healthy volunteers aged between 20 and 73 years over 5 anteroposterior electroencephalography scalp derivations. Spindle density, amplitude, and duration were higher in young subjects than in middle-aged and elderly subjects in all derivations, but the topography of age effects differed drastically. Age-related decline in density and amplitude was more prominent in anterior derivations, whereas duration showed a posterior prominence. Age groups did not differ in all-night spindle frequency for any derivation. These results show that age-related changes in sleep spindles follow distinct topographical patterns that are specific to each spindle characteristic. This topographical specificity may provide a useful biomarker to localize age-sensitive changes in underlying neural systems during normal and pathological aging.
Collapse
Affiliation(s)
- Nicolas Martin
- Department of Psychology, University of Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Forcelli PA, Janssen MJ, Vicini S, Gale K. Neonatal exposure to antiepileptic drugs disrupts striatal synaptic development. Ann Neurol 2012; 72:363-72. [PMID: 22581672 DOI: 10.1002/ana.23600] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 03/05/2012] [Accepted: 03/23/2012] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Drug exposure during critical periods of brain development may adversely affect nervous system function, posing a challenge for treating infants. This is of particular concern for treating neonatal seizures, as early life exposure to drugs such as phenobarbital is associated with adverse neurological outcomes in patients and induction of neuronal apoptosis in animal models. The functional significance of the preclinical neurotoxicity has been questioned due to the absence of evidence for functional impairment associated with drug-induced developmental apoptosis. METHODS We used patch-clamp recordings to examine functional synaptic maturation in striatal medium spiny neurons from neonatal rats exposed to antiepileptic drugs with proapoptotic action (phenobarbital, phenytoin, lamotrigine) and without proapoptotic action (levetiracetam). Phenobarbital-exposed rats were also assessed for reversal learning at weaning. RESULTS Recordings from control animals revealed increased inhibitory and excitatory synaptic connectivity between postnatal day (P)10 and P18. This maturation was absent in rats exposed at P7 to a single dose of phenobarbital, phenytoin, or lamotrigine. Additionally, phenobarbital exposure impaired striatal-mediated behavior on P25. Neuroprotective pretreatment with melatonin, which prevents drug-induced neurodevelopmental apoptosis, prevented the drug-induced disruption in maturation. Levetiracetam was found not to disrupt synaptic development. INTERPRETATION Our results provide the first evidence that exposure to antiepileptic drugs during a sensitive postnatal period impairs physiological maturation of synapses in neurons that survive the initial drug insult. These findings suggest a mechanism by which early life exposure to antiepileptic drugs can impact cognitive and behavioral outcomes, underscoring the need to identify therapies that control seizures without compromising synaptic maturation.
Collapse
Affiliation(s)
- Patrick A Forcelli
- Interdisciplinary Program in Neuroscience, Georgetown University, School of Medicine, Washington, DC, USA.
| | | | | | | |
Collapse
|
27
|
Kozlov AP, Nizhnikov ME, Varlinskaya EI, Spear NE. The role of social isolation in ethanol effects on the preweanling rat. Behav Brain Res 2012; 227:43-57. [PMID: 22051944 DOI: 10.1016/j.bbr.2011.10.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/19/2011] [Indexed: 01/08/2023]
Abstract
The present experiments investigated the effects of acute ethanol exposure on voluntary intake of 0.1% saccharin or water as well as behavioral and nociceptive reactivity in 12-day-old (P12) rats exposed to differing levels of isolation. The effects of ethanol emerged only during short-term social isolation (STSI) with different patterns observed in males and females and in pups exposed to saccharin or water. The 0.5g/kg ethanol dose selectively increased saccharin intake in females, decreased rearing activity in males and attenuated isolation-induced analgesia (IIA) in all water-exposed pups. Ingestion of saccharin decreased IIA, and the 0.5g/kg ethanol dose further reduced IIA. The 1.0g/kg ethanol dose, administered either intragastrically or intraparentionally, also decreased IIA in P12 females, but not in P9 pups. A significant correlation between voluntary saccharin intake and baseline nociceptive reactivity was revealed in saline injected animals, saccharin intake was inversely correlated with behavioral activation and latency of reaction to noxious heat after 0.5g/kg ethanol in females. The 0.5g/kg ethanol dose did not affect plasma corticosterone (CORT) measured 5h after maternal separation or 20min after ethanol injection. Female pups CORT level was inversely correlated with magnitude of IIA that accompanied the first episode of STSI (pretest isolation) 1.5-2h before CORT measurement. The present findings suggest that the anxiolytic properties of ethanol are responsible for enhancement of saccharin intake during STSI. Furthermore, differential reactivity of P12 males and females to STSI plays an important role in ethanol effects observed at this age.
Collapse
Affiliation(s)
- Andrey P Kozlov
- Center for Development & Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | | | | | | |
Collapse
|
28
|
Altered GABA signaling in early life epilepsies. Neural Plast 2011; 2011:527605. [PMID: 21826277 PMCID: PMC3150203 DOI: 10.1155/2011/527605] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 05/04/2011] [Accepted: 05/27/2011] [Indexed: 01/13/2023] Open
Abstract
The incidence of seizures is particularly high in the early ages of life. The immaturity of inhibitory systems, such as GABA, during normal brain development and its further dysregulation under pathological conditions that predispose to seizures have been speculated to play a major role in facilitating seizures. Seizures can further impair or disrupt GABAA signaling by reshuffling the subunit composition of its receptors or causing aberrant reappearance of depolarizing or hyperpolarizing GABAA receptor currents. Such effects may not result in epileptogenesis as frequently as they do in adults. Given the central role of GABAA signaling in brain function and development, perturbation of its physiological role may interfere with neuronal morphology, differentiation, and connectivity, manifesting as cognitive or neurodevelopmental deficits. The current GABAergic antiepileptic drugs, while often effective for adults, are not always capable of stopping seizures and preventing their sequelae in neonates. Recent studies have explored the therapeutic potential of chloride cotransporter inhibitors, such as bumetanide, as adjunctive therapies of neonatal seizures. However, more needs to be known so as to develop therapies capable of stopping seizures while preserving the age- and sex-appropriate development of the brain.
Collapse
|
29
|
Differential short-term plasticity at convergent inhibitory synapses to the substantia nigra pars reticulata. J Neurosci 2010; 30:14854-61. [PMID: 21048144 DOI: 10.1523/jneurosci.3895-10.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inhibitory projections from the striatum and globus pallidus converge onto GABAergic projection neurons of the substantia nigra pars reticulata (SNr). Based on existing structural and functional evidence, these pathways are likely to differentially regulate the firing of SNr neurons. We sought to investigate the functional differences in inhibitory striatonigral and pallidonigral traffic using whole-cell voltage clamp in brain slices with these pathways preserved. We found that striatonigral IPSCs exhibited a high degree of paired-pulse facilitation. We tracked this facilitation over development and found the facilitation as the animal aged, but stabilized by postnatal day 17 (P17), with a paired pulse ratio of 2. We also found that the recovery from facilitation accelerated over development, again, reaching a stable phenotype by P17. In contrast, pallidonigral synapses show paired-pulse depression, and this depression could be solely explained by presynaptic changes. The mean paired-pulse ratio of 0.67 did not change over development, but the recovery from depression slowed over development. Pallidonigral IPSCs were significantly faster than striatonigral IPSCs when measured at the soma. Finally, under current clamp, prolonged bursts of striatal IPSPs were able to consistently silence the pacemaker activity of nigral neurons, whereas pallidal inputs depressed, allowing nigral neurons to reinstate firing. These findings highlight the importance of differential dynamics of neurotransmitter release in regulating the circuit behavior of the basal ganglia.
Collapse
|
30
|
Huang X, Zhang H, Yang J, Wu J, McMahon J, Lin Y, Cao Z, Gruenthal M, Huang Y. Pharmacological inhibition of the mammalian target of rapamycin pathway suppresses acquired epilepsy. Neurobiol Dis 2010; 40:193-9. [PMID: 20566381 DOI: 10.1016/j.nbd.2010.05.024] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 05/11/2010] [Accepted: 05/17/2010] [Indexed: 10/19/2022] Open
Abstract
Inhibition of mTOR by rapamycin has been shown to suppress seizures in TSC/PTEN genetic models. Rapamycin, when applied immediately before or after a neurological insult, also prevents the development of spontaneous recurrent seizures (epileptogenesis) in an acquired model. In the present study, we examined the mTOR pathway in rats that had already developed chronic spontaneous seizures in a pilocarpine model. We found that mTOR is aberrantly activated in brain tissues from rats with chronic seizures. Furthermore, inhibition of mTOR by rapamycin treatment significantly reduces seizure activity. Finally, mTOR inhibition also significantly suppresses mossy fiber sprouting. Our findings suggest the possibility for a much broader window for intervention for some acquired epilepsies by targeting the mTOR pathway.
Collapse
Affiliation(s)
- Xiaoxing Huang
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
King BM. Point: a call for proper usage of "gender" and "sex" in biomedical publications. Am J Physiol Regul Integr Comp Physiol 2010; 298:R1700-1. [PMID: 20357018 DOI: 10.1152/ajpregu.00694.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Bruce M King
- Department of Psychology, Clemson University, Clemson, South Carolina 29634, USA.
| |
Collapse
|
32
|
Galanopoulou AS. Mutations affecting GABAergic signaling in seizures and epilepsy. Pflugers Arch 2010; 460:505-23. [PMID: 20352446 DOI: 10.1007/s00424-010-0816-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 02/18/2010] [Accepted: 02/23/2010] [Indexed: 02/02/2023]
Abstract
The causes of epilepsies and epileptic seizures are multifactorial. Genetic predisposition may contribute in certain types of epilepsies and seizures, whether idiopathic or symptomatic of genetic origin. Although these are not very common, they have offered a unique opportunity to investigate the molecular mechanisms underlying epileptogenesis and ictogenesis. Among the implicated gene mutations, a number of GABAA receptor subunit mutations have been recently identified that contribute to several idiopathic epilepsies, febrile seizures, and rarely to certain types of symptomatic epilepsies, like the severe myoclonic epilepsy of infancy. Deletion of GABAA receptor genes has also been linked to Angelman syndrome. Furthermore, mutations of proteins controlling chloride homeostasis, which indirectly defines the functional consequences of GABAA signaling, have been identified. These include the chloride channel 2 (CLCN2) and the potassium chloride cotransporter KCC3. The pathogenic role of CLCN2 mutations has not been clearly demonstrated and may represent either susceptibility genes or, in certain cases, innocuous polymorphisms. KCC3 mutations have been associated with hereditary motor and sensory polyneuropathy with corpus callosum agenesis (Andermann syndrome) that often manifests with epileptic seizures. This review summarizes the recent progress in the genetic linkages of epilepsies and seizures to the above genes and discusses potential pathogenic mechanisms that contribute to the age, sex, and conditional expression of these seizures in carriers of these mutations.
Collapse
Affiliation(s)
- Aristea S Galanopoulou
- Saul R. Korey Department of Neurology and Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Kennedy Center Room 306, Bronx, NY 10461, USA.
| |
Collapse
|