1
|
Fatahi N, Jafari-Sabet M, Vahabzadeh G, Komaki A. Role of hippocampal and prefrontal cortical cholinergic transmission in combination therapy valproate and cannabidiol in memory consolidation in rats: involvement of CREB- BDNF signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5029-5047. [PMID: 38189934 DOI: 10.1007/s00210-023-02941-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
PURPOSE Cognitive disorders are associated with valproate and drugs used to treat neuropsychological diseases. Cannabidiol (CBD) has beneficial effects on cognitive function. This study examined the effects of co-administration of CBD and valproate on memory consolidation, cholinergic transmission, and cyclic AMP response element-binding protein (CREB)-brain-derived neurotrophic factor (BDNF) signaling pathway in the prefrontal cortex (PFC) and hippocampus (HPC). METHODS One-trial, step-through inhibitory test was used to evaluate memory consolidation in rats. The intra-CA1 injection of physostigmine and atropine was performed to assess the role of cholinergic transmission in this co-administration. Phosphorylated CREB (p-CREB)/CREB ratio and BDNF levels in the PFC and HPC were evaluated. RESULTS Post-training intraperitoneal (i.p.) valproate injection reduced memory consolidation; however, post-training co-administration of CBD with valproate ameliorated memory impairment induced by valproate. Post-training intra-CA1 injection of physostigmine at the ineffective doses in memory consolidation (0.5 and 1 µg/rat), plus injection of 10 mg/kg of CBD as an ineffective dose, improved memory loss induced by valproate, which was associated with BDNF and p-CREB level enhancement in the PFC and HPC. Conversely, post-training intra-CA1 injection of ineffective doses of atropine (1 and 2 µg/rat) reduced the positive effects of injection of CBD at a dose of 20 mg/kg on valproate-induced memory loss associated with BDNF and p-CREB level reduction in the PFC and HPC. CONCLUSION The results indicated a beneficial interplay between valproate and CBD in the process of memory consolidation, which probably creates this interaction through the BDNF-CREB signaling pathways in the cholinergic transmission of the PFC and HPC regions.
Collapse
Affiliation(s)
- Navid Fatahi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Majid Jafari-Sabet
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Gelareh Vahabzadeh
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
2
|
Wang M, Xu B, Xie Y, Yao G, Chen Y. Mir155hg Accelerates Hippocampal Neuron Injury in Convulsive Status Epilepticus by Inhibiting Microglial Phagocytosis. Neurochem Res 2024; 49:1782-1793. [PMID: 38555337 DOI: 10.1007/s11064-024-04131-x] [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: 11/22/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 04/02/2024]
Abstract
Convulsive status epilepticus (CSE) is a common critical neurological condition that can lead to irreversible hippocampal neuron damage and cognitive dysfunction. Multiple studies have demonstrated the critical roles that long non-coding RNA Mir155hg plays in a variety of diseases. However, less is known about the function and mechanism of Mir155hg in CSE. Here we investigate and elucidate the mechanism underlying the contribution of Mir155hg to CSE-induced hippocampal neuron injury. By applying high-throughput sequencing, we examined the expression of differentially expressed genes in normal and CSE rats. Subsequent RT-qPCR enabled us to measure the level of Mir155hg in rat hippocampal tissue. Targeted knockdown of Mir155hg was achieved by the AAV9 virus. Additionally, we utilized HE and Tunel staining to evaluate neuronal injury. Immunofluorescence (IF), Golgi staining, and brain path clamping were also used to detect the synaptic plasticity of hippocampal neurons. Finally, through IF staining and Sholl analysis, we assessed the degree of microglial phagocytic function. It was found that the expression of Mir155hg was elevated in CSE rats. HE and Tunel staining results showed that Mir155hg knockdown suppressed the hippocampal neuron loss and apoptosis followed CSE. IF, Golgi staining and brain path clamp data found that Mir155hg knockdown enhanced neuronal synaptic plasticity. The results from IF staining and Sholl analysis showed that Mir155hg knockdown enhanced microglial phagocytosis. Our findings suggest that Mir155hg promotes CSE-induced hippocampal neuron injury by inhibiting microglial phagocytosis.
Collapse
Affiliation(s)
- Ming Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Binyuan Xu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yangmei Xie
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ge Yao
- Department of Neurology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Yinghui Chen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
3
|
Jia Y, Tang L, Yao Y, Zhuo L, Qu D, Chen X, Ji Y, Tao J, Zhu Y. Low-intensity exercise combined with sodium valproate attenuates kainic acid-induced seizures and associated co-morbidities by inhibiting NF-κB signaling in mice. Front Neurol 2022; 13:993405. [PMID: 36212646 PMCID: PMC9534325 DOI: 10.3389/fneur.2022.993405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Sodium valproate (VPA) is a broad-spectrum anticonvulsant that is effective both in adults and children suffering from epilepsy, but it causes psychiatric and behavioral side effects in patients with epilepsy. In addition, 30% of patients with epilepsy develop resistance to VPA. At present, regular physical exercise has shown many benefits and has become an effective complementary therapy for various brain diseases, including epilepsy. Therefore, we wondered whether VPA combined with exercise would be more effective in the treatment of seizures and associated co-morbidities. Here, we used a mouse model with kainic acid (KA)-induced epilepsy to compare the seizure status and the levels of related co-morbidities, such as cognition, depression, anxiety, and movement disorders, in each group using animal behavioral experiment and local field potential recordings. Subsequently, we investigated the mechanism behind this phenomenon by immunological means. Our results showed that low-intensity exercise combined with VPA reduced seizures and associated co-morbidities. This phenomenon seems to be related to the Toll-like receptor 4, activation of the nuclear factor kappa B (NF-κB), and release of interleukin 1β (IL-1β), tumor necrosis factor α (TNF-α), and IL-6. In brief, low-intensity exercise combined with VPA enhanced the downregulation of NF-κB-related inflammatory response, thereby alleviating the seizures, and associated co-morbidities.
Collapse
Affiliation(s)
- Yuxiang Jia
- School of Medicine, Shanghai University, Shanghai, China
| | - Lele Tang
- Department of Neurology and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Yao
- School of Medicine, Shanghai University, Shanghai, China
| | - Limin Zhuo
- School of Medicine, Shanghai University, Shanghai, China
| | - Dongxiao Qu
- Department of Neurology and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xingxing Chen
- Department of Neurology and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghua Ji
- School of Medicine, Shanghai University, Shanghai, China
- *Correspondence: Yonghua Ji
| | - Jie Tao
- Department of Neurology and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Jie Tao
| | - Yudan Zhu
- Department of Neurology and Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Yudan Zhu
| |
Collapse
|
4
|
Shavit-Stein E, Berkowitz S, Davidy T, Fennig U, Gofrit SG, Dori A, Maggio N. Modulation of the Thrombin Pathway Restores LTP in a Pilocarpine Mice Model of Status Epilepticus. Front Cell Neurosci 2022; 16:900925. [PMID: 35685989 PMCID: PMC9170943 DOI: 10.3389/fncel.2022.900925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/02/2022] [Indexed: 11/28/2022] Open
Abstract
Background Status epilepticus (SE) leads to memory impairment following a seizure, attributed to long-term potentiation (LTP) reduction. Thrombin, a coagulation factor that activates protease-activated receptor 1 (PAR1) is involved in cognitive impairment following traumatic brain injury by reducing hippocampal LTP and in seizures as seen in a SE pilocarpine-induced mice model. Thrombin pathway inhibition prevents this cognitive impairment. We evaluated the effect of thrombin pathway inhibition in the pilocarpine-induced SE mice model, on LTP, hippocampal, and serum markers for inflammation, the PAR1 pathway, and neuronal cell damage. Methods SE was induced by injecting C57BL/6J mice with pilocarpine. Before pilocarpine injection, mice were injected with either the specific thrombin inhibitor α-NAPAP [Nα-(2-naphthalene-sulfonylglycyl)-4-amidino-DL-phenylalaninepiperidide], the PAR1 antagonist SCH79797, or vehicle-only solution. Recordings of excitatory postsynaptic potentials (EPSP) were conducted from hippocampal slices 24 h following pilocarpine injection. Hippocampal real-time PCR for the quantification of the PAR1, prothrombin, and tumor necrosis factor α (TNF-α) mRNA expression levels was conducted. Serum levels of neurofilament light chain (NfL) and TNF-α were measured by a single molecule array assay. Results The EPSP was reduced in the pilocarpine-induced SE mice (p < 0.001). This reduction was prevented by both NAPAP and SCH79797 treatments (p < 0.001 for both treatments). Hippocampal expression of TNF-α was elevated in the pilocarpine-induced SE group compared to the control (p < 0.01), however, serum levels of TNF-α were not changed. NfL levels were elevated in the pilocarpine-induced SE group (p = 0.04) but not in the treated groups. Conclusions Pilocarpine-induced SE reduces LTP, in a thrombin PAR1-related mechanism. Elevation of serum NfL supports neuronal damage accompanying this functional abnormality which may be prevented by PAR1 pathway modulation.
Collapse
Affiliation(s)
- Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The TELEM Rubin Excellence in Biomedical Research Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Davidy
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Uri Fennig
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Shani Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Amir Dori
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Nicola Maggio
| |
Collapse
|
5
|
Raja K, Prabahar A, Arputhanatham SS. A Simple Computational Approach to Identify Potential Drugs for Multiple Sclerosis and Cognitive Disorders from Expert Curated Resources. Methods Mol Biol 2022; 2496:111-121. [PMID: 35713861 DOI: 10.1007/978-1-0716-2305-3_6] [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] [Indexed: 06/15/2023]
Abstract
Multiple sclerosis, a disease of central nervous system leads to potential disability. In the USA, one million cases are diagnosed with multiple sclerosis in 2019. Multiple sclerosis is identified as one of the diseases causing global burden. Cognitive disorder is highly prevalent among 43-70% of multiple sclerosis patients. However, treating cognitive disorder in multiple sclerosis patients is mostly ignored and this leads to several complications. We utilized various expert curated resources to identify potential drugs for multiple sclerosis and cognitive disorder, with specific focus on identifying drugs that are capable of treating both the conditions. We used simple text mining techniques to compile two databases, disease-drug association database and gene-drug interaction database from various existing standard resources. Our study suggests four drugs, Baclofen, Levodopa, Minocycline, and Vitamin B12, for treating both multiple sclerosis and cognitive disorder. In addition, our approach suggests six drugs for multiple sclerosis and 10 drugs for cognitive disorder. We obtained pharmacologist opinion on the drugs suggested for each condition and provided literature evidence for our claim. Here, we present our computational approach as a protocol such that it can be applied to other comorbid diseases that did not gain much attention so far.
Collapse
Affiliation(s)
- Kalpana Raja
- Regenerative Biology, The Morgridge Institute for Research, Madison, WI, USA.
| | - Archana Prabahar
- R&D Division, Eriks-Precision Components India Pvt Ltd, Mohali, Punjab, India
| | | |
Collapse
|
6
|
Ghamkharinejad G, Marashi SH, Foolad F, Javan M, Fathollahi Y. Unconditioned and learned morphine tolerance influence hippocampal-dependent short-term memory and the subjacent expression of GABA-A receptor alpha subunits. PLoS One 2021; 16:e0253902. [PMID: 34500453 PMCID: PMC8428970 DOI: 10.1371/journal.pone.0253902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 06/15/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND ɣ-aminobutyric acid (GABA) facilitator valproic acid may be able to curb memory disruption induced by morphine exposure. OBJECTIVE The effects of the GABA facilitator valproic acid on the behavioral tolerance induced by morphine were investigated. Then hippocampal-dependent tasks named spatial-working and short-term memory procedures using the Y-maze apparatus were examined in morphine tolerant rats. Finally, the changes in the expression of hippocampal GABA-A receptors underlying morphine tolerance were also examined. METHODS Rats were treated with daily morphine injections, with or without distinct contextual pairing. To examine the effect of valproic acid on morphine tolerance expression, valproic acid was pretreated an hour before morphine. Spatial-working and short-term memory procedures using the Y-maze apparatus were examined in morphine tolerant rats. Afterwards the changes in the expression of hippocampal GABAα receptors using the quantitative real-time PCR and western blot techniques to detect GABArα subunits mRNAs and protein level were studied. RESULTS Our results showed that both learned and non-associative morphine tolerance influence short-term memory and the subjacent expression of GABArα mRNAs and protein level. Despite its attenuating effects on the development and expression of both learned and non-associative morphine tolerance, only associative morphine tolerance-induced memory dysfunction was ameliorated by valproic acid pretreatment. We also found that the expression of GABArα1, α2, α5 subunits mRNAs and GABAα protein level were affected heavier in associative morphine tolerant rats. CONCLUSION Our data supports the hypothesis that unconditioned and learned morphine tolerance influences short-term memory and the expression of GABArα 1, α2, α5 mRNAs and GABArα protein level differently, and adds to our understanding of the behavioral and molecular aspects of the learned tolerance to morphine effects.
Collapse
Affiliation(s)
- Ghazaleh Ghamkharinejad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Hossein Marashi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Forough Foolad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
7
|
Vyas P, Tulsawani RK, Vohora D. Loss of Protection by Antiepileptic Drugs in Lipopolysaccharide-primed Pilocarpine-induced Status Epilepticus is Mediated via Inflammatory Signalling. Neuroscience 2020; 442:1-16. [PMID: 32592825 DOI: 10.1016/j.neuroscience.2020.06.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/22/2022]
Abstract
The evidences from various studies show the association of peripheral and neuronal inflammation with complex pathophysiology of status epilepticus (SE). In this view, the present work attempted to develop a model of neuronal inflammation mediated SE by combining both epileptic and inflammatory components of the disease and also to mimic SE co-morbid with systemic inflammation by peripheral administration of the lipopolysaccharide (LPS) 2 h prior to the pilocarpine (PILO) induction in C57BL/6 mice. We evaluated the anti-convulsant and neuroprotective effects of 7-day prophylactic treatment with three conventional anti-epileptic drugs (Sodium valproate, SVP 300 mg/kg p.o.; Carbamazepine CBZ 100 mg/kg p.o.; Levetiracetam; LEV 200 mg/kg p.o.) of widespread clinical use. Morris water maze and Rota rod tests were carried out 24-h post-exposure to evaluate the neurobehavioral co-morbidities associated with neuroinflammation-mediated status epilepticus. Upon priming with LPS, the loss of protection against PILO-induced seizures was observed by SVP and CBZ, however, LEV showed protection by delaying the seizures. Dramatic elevation in the seizure severity and neuronal loss demonstrated the possible pro-convulsant effect of LPS in the PILO model. Also, the decreased cytokine levels by the AEDs showed their association with NF-κB, IL-1β, IL-6, TNF-α and TGF-β pathways in PILO model. The loss of protective activities of SVP and CBZ in LPS+PILO model was due to increased cytokine levels associated with over-activation of neuroinflammatory pathways, however, partial efficacy of LEV is possibly due to association of other neuroinflammatory mechanisms. The current work provides direct evidence of the contribution of increased peripheral and neuronal inflammation in seizures via regulation of inflammatory pathways in the brain.
Collapse
Affiliation(s)
- Preeti Vyas
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Raj Kumar Tulsawani
- Defense Institute of Physiology & Allied Science, Defense Research and Development Organization, New Delhi, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| |
Collapse
|
8
|
Meenu M, Reeta KH, Dinda AK, Kottarath SK, Gupta YK. Evaluation of sodium valproate loaded nanoparticles in acute and chronic pentylenetetrazole induced seizure models. Epilepsy Res 2019; 158:106219. [PMID: 31726286 DOI: 10.1016/j.eplepsyres.2019.106219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/27/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Efficacy of sodium valproate in epilepsy is limited by its poor blood brain barrier penetration and side effects. Nanoparticles may offer a better drug delivery system to overcome these limitations. This study evaluated the efficacy of sodium valproate encapsulated in nanoparticles in pentylenetetrazole (PTZ) induced acute and kindling models of seizures in male Wistar rats. METHODS Poly lactic-co-glycolic acid (PLGA) based, polysorbate 80 stabilized sodium valproate loaded nanoparticles (nano sodium valproate) and rhodamine loaded nanoparticles (RLN) were formulated by double emulsion- solvent evaporation method and characterized for their size, shape, zeta potential and drug loading percentage. RLN was used to demonstrate blood brain barrier (BBB) permeability of nanoparticles. Serum drug levels were estimated using high performance liquid chromatography. The efficacy of standard sodium valproate (300 mg/kg) and nano sodium valproate (∼300, ∼150 and ∼75 mg/kg of sodium valproate) were evaluated in experimental animal models of seizures along with their effects on behavioral and oxidative stress parameters. Drugs were administered 60 min before PTZ in acute model. In the kindling model, drugs were administered every day while PTZ was administered on alternate days 60 min after drug administration. All the study drugs/compounds were administered intraperitoneally. RESULTS RLN were observed to be clustered in cortex which implied that the nanoparticles crossed BBB. Both standard sodium valproate and nano sodium valproate reached therapeutic serum level at 15 min and 1 h, but were undetectable in serum at 24 h. In acute PTZ (60 mg/kg) model, nano sodium valproate (∼300 mg/kg of sodium valproate) and standard sodium valproate showed protection against seizures till 6 h and 4 h, respectively. There were significant behavioral impairment and oxidative stress with standard sodium valproate in acute model as compared to nano sodium valproate at 6 h. In kindling model, induced with PTZ (30 mg/kg, every alternate day for 42 days), complete protection from seizures was observed with nano sodium valproate (∼150 mg/kg and ∼75 mg/kg of sodium valproate) and standard sodium valproate (300 mg/kg). Similarly, significant protection from behavioral impairment and oxidative stress was observed with standard sodium valproate and nano sodium valproate as compared to PTZ. CONCLUSION When compared to conventional therapy, nano sodium valproate showed protection from seizures at reduced doses and for a longer duration in animal models of epilepsy. This study suggests the potential of nano sodium valproate in the treatment of epilepsy.
Collapse
Affiliation(s)
- Meenakshi Meenu
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| | - Amit Kumar Dinda
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Yogendra Kumar Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
9
|
Wang MJ, Jiang L, Chen HS, Cheng L. Levetiracetam Protects Against Cognitive Impairment of Subthreshold Convulsant Discharge Model Rats by Activating Protein Kinase C (PKC)-Growth-Associated Protein 43 (GAP-43)-Calmodulin-Dependent Protein Kinase (CaMK) Signal Transduction Pathway. Med Sci Monit 2019; 25:4627-4638. [PMID: 31266934 PMCID: PMC6601366 DOI: 10.12659/msm.913542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Subclinical epileptiform discharges (SEDs) are defined as epileptiform electroencephalographic (EEG) discharges without clinical signs of seizure in patients. The subthreshold convulsant discharge (SCD) is a frequently used model for SEDs. This study aimed to investigate the effect of levetiracetam (LEV), an anti-convulsant drug, on cognitive impairment of SCD model rats and to assess the associated mechanisms. Material/Methods A SCD rat model was established. Rats were divided into an SCD group, an SCD+ sodium valproate (VPA) group, and an SCD+ levetiracetam (LEV) group. The Morris water maze was used to evaluate the capacity of positioning navigation and space exploration. The field excitatory post-synaptic potentials (fEPSPs) were evaluated using a bipolar stimulation electrode. NCAM, GAP43, PS95, and CaMK II levels were detected using Western blot and RT-PCR, respectively. PKC activity was examined by a non-radioactive method. Results LEV shortens the latency of platform seeking in SCD rats in positioning navigation. fEPSP slopes were significantly lower in the SCD group, and LEV treatment significantly enhanced the fEPSP slopes compared to the SCD group (P<0.05). The NCAM and GAP-43 levels were increased and PSD-95 levels were increased in SCD rats (P<0.05), which were improved by LEV treatment. The PKC activity and CaMK II levels were decreased in SCD rats and LEV treatment significantly enhanced PKC activity and increased CaMK II levels. Conclusions Cognitive impairment in of SCD model rats may be caused by decreased PKC activity, low expression of CaMK II, and inhibition of LTP formation. LEV can improve cognitive function by activating the PKC-GAP-43-CaMK signal transduction pathway.
Collapse
Affiliation(s)
- Min-Jian Wang
- Department of Psychology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland).,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China (mainland).,Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China (mainland)
| | - Li Jiang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China (mainland).,Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China (mainland).,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Heng-Sheng Chen
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Li Cheng
- Key Laboratory of Pediatrics in Chongqing, Chongqing, China (mainland).,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
10
|
Wu P, Hu Y, Li XJ, Cheng M, Jiang L. Sodium valproate suppresses abnormal neurogenesis induced by convulsive status epilepticus. Neural Regen Res 2018; 14:480-484. [PMID: 30539816 PMCID: PMC6334603 DOI: 10.4103/1673-5374.245475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Status epilepticus has been shown to activate the proliferation of neural stem cells in the hippocampus of the brain, while also causing a large amount of neuronal death, especially in the subgranular zone of the dentate gyrus and the subventricular zone. Simultaneously, proliferating stem cells tend to migrate to areas with obvious damage. Our previous studies have clearly confirmed the effect of sodium valproate on cognitive function in rats with convulsive status epilepticus. However, whether neurogenesis can play a role in the antiepileptic effect of sodium valproate remains unknown. A model of convulsive status epilepticus was established in Wistar rats by intraperitoneal injection of 3 mEq/kg lithium chloride, and intraperitoneal injection of pilocarpine 40 mg/kg after 18–20 hours. Sodium valproate (100, 200, 300, 400, 500, or 600 mg/kg) was intragastrically administered six times every day (4-hour intervals) for 5 days. To determine the best dosage, sodium valproate concentration was measured from the plasma. The effective concentration of sodium valproate in the plasma of the rats that received the 300-mg/kg intervention was 82.26 ± 11.23 μg/mL. Thus, 300 mg/kg was subsequently used as the intervention concentration of sodium valproate. The following changes were seen: Recording excitatory postsynaptic potentials in the CA1 region revealed high-frequency stimulation-induced long-term potentiation. Immunohistochemical staining for BrdU-positive cells in the brain revealed that sodium valproate intervention markedly increased the success rate and the duration of induced long-term potentiation in rats with convulsive status epilepticus. The intervention also reduced the number of newborn neurons in the subgranular area of the hippocampus and subventricular zone and inhibited the migration of newborn neurons to the dentate gyrus. These results indicate that sodium valproate can effectively inhibit the abnormal proliferation and migration of neural stem cells and newborn neurons after convulsive status epilepticus, and improve learning and memory ability.
Collapse
Affiliation(s)
- Peng Wu
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Hu
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiu-Juan Li
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Min Cheng
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Li Jiang
- Department of Neurology, Children's Hospital of Chongqing Medical University; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Key Laboratory of Pediatrics in Chongqing, Chongqing, China
| |
Collapse
|
11
|
Xu MY, Wong AHC. GABAergic inhibitory neurons as therapeutic targets for cognitive impairment in schizophrenia. Acta Pharmacol Sin 2018; 39:733-753. [PMID: 29565038 DOI: 10.1038/aps.2017.172] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/25/2017] [Indexed: 12/24/2022] Open
Abstract
Schizophrenia is considered primarily as a cognitive disorder. However, functional outcomes in schizophrenia are limited by the lack of effective pharmacological and psychosocial interventions for cognitive impairment. GABA (gamma-aminobutyric acid) interneurons are the main inhibitory neurons in the central nervous system (CNS), and they play a critical role in a variety of pathophysiological processes including modulation of cortical and hippocampal neural circuitry and activity, cognitive function-related neural oscillations (eg, gamma oscillations) and information integration and processing. Dysfunctional GABA interneuron activity can disrupt the excitatory/inhibitory (E/I) balance in the cortex, which could represent a core pathophysiological mechanism underlying cognitive dysfunction in schizophrenia. Recent research suggests that selective modulation of the GABAergic system is a promising intervention for the treatment of schizophrenia-associated cognitive defects. In this review, we summarized evidence from postmortem and animal studies for abnormal GABAergic neurotransmission in schizophrenia, and how altered GABA interneurons could disrupt neuronal oscillations. Next, we systemically reviewed a variety of up-to-date subtype-selective agonists, antagonists, positive and negative allosteric modulators (including dual allosteric modulators) for α5/α3/α2 GABAA and GABAB receptors, and summarized their pro-cognitive effects in animal behavioral tests and clinical trials. Finally, we also discuss various representative histone deacetylases (HDAC) inhibitors that target GABA system through epigenetic modulations, GABA prodrug and presynaptic GABA transporter inhibitors. This review provides important information on current potential GABA-associated therapies and future insights for development of more effective treatments.
Collapse
|
12
|
Marsh J, Alifragis P. Synaptic dysfunction in Alzheimer's disease: the effects of amyloid beta on synaptic vesicle dynamics as a novel target for therapeutic intervention. Neural Regen Res 2018; 13:616-623. [PMID: 29722304 PMCID: PMC5950662 DOI: 10.4103/1673-5374.230276] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The most prevalent form of dementia in the elderly is Alzheimer's disease. A significant contributing factor to the progression of the disease appears to be the progressive accumulation of amyloid-β42 (Aβ42), a small hydrophobic peptide. Unfortunately, attempts to develop therapies targeting the accumulation of Aβ42 have not been successful to treat or even slow down the disease. It is possible that this failure is an indication that targeting downstream effects rather than the accumulation of the peptide itself might be a more effective approach. The accumulation of Aβ42 seems to affect various aspects of physiological cell functions. In this review, we provide an overview of the evidence that implicates Aβ42 in synaptic dysfunction, with a focus on how it contributes to defects in synaptic vesicle dynamics and neurotransmitter release. We discuss data that provide new insights on the Aβ42 induced pathology of Alzheimer's disease and a more detailed understanding of its contribution to the synaptic deficiencies that are associated with the early stages of the disease. Although the precise mechanisms that trigger synaptic dysfunction are still under investigation, the available data so far has enabled us to put forward a model that could be used as a guide to generate new therapeutic targets for pharmaceutical intervention.
Collapse
Affiliation(s)
- Jade Marsh
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | - Pavlos Alifragis
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, UK
| |
Collapse
|
13
|
Xu Y, Li Z, Yao L, Zhang X, Gan D, Jiang M, Wang N, Chen G, Wang X. Altered Norbin Expression in Patients with Epilepsy and a Rat Model. Sci Rep 2017; 7:13970. [PMID: 29070854 PMCID: PMC5656659 DOI: 10.1038/s41598-017-13248-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 09/05/2017] [Indexed: 01/17/2023] Open
Abstract
Norbin is widely distributed in neuronal tissues, is a regulator of Ca2+/calmodulin-dependent protein kinase II (CaMKII) phosphorylation. Norbin is also an important endogenous modulator of metabotropic glutamate receptor 5 (mGluR5) signaling, and nervous system-specific homozygous gene disruptions, result in epileptic seizures. In this study, we aimed to investigate norbin expression patterns in epilepsy and to elucidate the relationships between norbin and mGluR5 and p-CaMKII in epilepsy. Double-immunolabeling, immunohistochemistry and immunoblotting studies showed that norbin was downregulated in the temporal neocortex of patients with temporal lobe epilepsy (TLE) compared with control subjects. Moreover, in a rat model of lithium chloride-pilocarpine-induced epilepsy, norbin expression began to decrease at 6 h after the onset of status epilepticus and remained at a low level until 60 days. In addition, p-CaMKII expression was significantly increased in both patients with TLE and in animal model. Norbin and mGluR5 were found to be co-expressed in neurons of epileptic tissues. Finally, norbin over-expression facilitated by injections of adeno-associated viral vector into the rat hippocampus increased latency and survival in the lithium chloride-pilocarpine model. Thus, our results indicate norbin participates in the pathogenesis of epilepsy, perhaps by modulating mGluR5 signaling, regulating CaMKII phosphorylation, and may exert antiepileptic effects.
Collapse
Affiliation(s)
- Yali Xu
- Department of Geriatrics, Chongqing General Hospital, 104 Pipashan Street, Chongqing, China.,Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 1 Youyi Road, Chongqing, China
| | - Zengyou Li
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 1 Youyi Road, Chongqing, China
| | - Li Yao
- Health Checkup Center, Chongqing General Hospital, 104 Pipashan Street, Chongqing, China
| | - Xingping Zhang
- Department of Geriatrics, Chongqing General Hospital, 104 Pipashan Street, Chongqing, China
| | - Dan Gan
- Department of Geriatrics, Chongqing General Hospital, 104 Pipashan Street, Chongqing, China
| | - Manchun Jiang
- Department of Geriatrics, Chongqing General Hospital, 104 Pipashan Street, Chongqing, China
| | - Na Wang
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 1 Youyi Road, Chongqing, China
| | - Guojun Chen
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 1 Youyi Road, Chongqing, China
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, 1 Youyi Road, Chongqing, China.
| |
Collapse
|