1
|
Uliana DL, Martinez A, Grace AA. THPP-1 PDE10A inhibitor reverses the cognitive deficits and hyperdopaminergic state in a neurodevelopment model of schizophrenia. Schizophr Res 2024; 274:315-326. [PMID: 39437478 DOI: 10.1016/j.schres.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/26/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Schizophrenia (SCZ) is a complex neuropsychiatric disorder characterized by positive, negative, and cognitive symptoms. The neurodevelopmental methylazoxy-methanol acetate (MAM) rodent model replicates key neurobiological features of SCZ which includes hyperdopaminergic states in the ventral tegmental area (VTA) and cognitive deficits. Typical and atypical antipsychotics are primarily effective in treating the positive symptoms of SCZ but often fall short of addressing cognitive deficits. A promising therapeutic approach for treating all symptoms of SCZ has emerged through the inhibition of phosphodiesterase 10 A (PDE10A). Our study aim was to investigate the impact of acute and chronic THPP-1 (PDE10A inhibitor) treatment, in MAM rats, focusing on cognitive deficits and VTA dopamine (DA) activity. Adult offspring of pregnant rats treated with Saline or MAM (20 mg/kg) on gestational day 17 were treated with THPP-1 acutely (male/female rats; 3 mg/kg) at postnatal day (PD) 70-80 or chronically (males; 3 weeks; 2-3 mg/kg) from PD 70-91 and tested in the novel object recognition test and electrophysiological recording of DA neurons in the VTA. Acute THPP-1 treatment reversed cognitive impairments and normalized the increased number of active DA neurons in the VTA of male and female MAM rats, without affecting control rats. Also, chronic THPP-1 treatment reversed cognitive deficits and normalized DA hyperactivity in the VTA of male MAM rats. The efficacy of THPP-1 in reversing MAM-induced impairments underscores its ability to target disease-specific circuitry without affecting normal regulated systems in control rats. Our findings highlight the therapeutic potential of THPP-1 for addressing cognitive deficits and DA dysregulation in SCZ.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Angela Martinez
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Wang M, Gu Y, Li Q, Feng B, Lv X, Zhang H, Kong Q, Dong Z, Tian X, Zhang Y. The Traf2 and NcK interacting kinase inhibitor NCB-0846 suppresses seizure activity involving the decrease of GRIA1. Genes Dis 2024; 11:100997. [PMID: 38292191 PMCID: PMC10826163 DOI: 10.1016/j.gendis.2023.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/09/2023] [Accepted: 03/29/2023] [Indexed: 02/01/2024] Open
Abstract
Epilepsy, one of the most common neurological disorders, is characterized by spontaneous recurrent seizures. Temporal lobe epilepsy (TLE) is one of the most common medically intractable seizure disorders. Traf2-and NcK-interacting kinase (TNIK) has recently attracted attention as a critical modulation target of many neurological and psychiatric disorders, but its role in epilepsy remains unclear. In this study, we hypothesized the involvement of TNIK in epilepsy and investigated TNIK expression in patients with intractable TLE and in a pilocarpine-induced rat model of epilepsy by western blotting, immunofluorescence, and immunohistochemistry. A pentylenetetrazole (PTZ)-induced epilepsy rat model was used to determine the effect of the TNIK inhibitor NCB-0846 on behavioral manifestations of epilepsy. Coimmunoprecipitation (Co-IP)/mass spectrometry (MS) was used to identify the potential mechanism. Through Co-IP, we detected and confirmed the main potential TNIK interactors. Subcellular fractionation was used to establish the effect of NCB-0846 on the expression of the main interactors in postsynaptic density (PSD) fractions. We found that TNIK was primarily located in neurons and decreased significantly in epilepsy model rats and TLE patients compared with controls. NCB-0846 delayed kindling progression and decreased seizure severity. Co-IP/MS identified 63 candidate TNIK interactors in rat hippocampi, notably CaMKII. Co-IP showed that TNIK might correlate with endogenous GRIA1, SYN2, PSD-95, CaMKIV, GABRG1, and GABRG2. In addition, the significant decrease in GRIA1 in hippocampal total lysate and PSDs after NCB-0846 treatment might help modify the progression of PTZ kindling. Our results suggest that TNIK contributes to epileptic pathology and is a potential antiepileptic drug target.
Collapse
Affiliation(s)
- Min Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Yixue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Qiubo Li
- Department of Pediatrics, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Bangzhe Feng
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Xinke Lv
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Hao Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Qingxia Kong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
| | - Zhifang Dong
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Yanke Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
3
|
Dell'Isola GB, Verrotti A, Sciaccaluga M, Dini G, Ferrara P, Parnetti L, Costa C. Cannabidiol: metabolism and clinical efficacy in epileptic patients. Expert Opin Drug Metab Toxicol 2024; 20:119-131. [PMID: 38465404 DOI: 10.1080/17425255.2024.2329733] [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: 12/12/2023] [Accepted: 03/08/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION The landscape of epilepsy treatment has undergone a significant transformation with the emergence of cannabidiol as a potential therapeutic agent. Epidiolex, a pharmaceutical formulation of highly purified CBD, garnered significant attention not just for its therapeutic potential but also for being the first cannabis-derived medication to obtain approval from regulatory bodies. AREA COVERED In this narrative review the authors explore the intricate landscape of CBD as an antiseizure medication, deepening into its pharmacological mechanisms and clinical trials involving various epileptic encephalopathies. This exploration serves as a comprehensive guide, shedding light on a compound that holds promise for individuals contending with the significant challenges of drug-resistant epilepsy. EXPERT OPINION Rigorous studies highlight cannabidiol's efficacy, safety profile, and potential cognitive benefits, warranting further exploration for its approval in various drug-resistant epilepsy forms. As a promising therapeutic option, cannabidiol not only demonstrates efficacy in seizure control but also holds the potential for broader enhancements in the quality of life, especially for patients with epileptic encephalopathies.
Collapse
Affiliation(s)
| | | | - Miriam Sciaccaluga
- Section of Neurology, Laboratory of Experimental Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- "Mauro Baschirotto" Institute for Rare Diseases - BIRD Foundation Onlus, Longare, Vicenza, Italy
| | - Gianluca Dini
- Department of Pediatrics, University of Perugia, Perugia, Italy
| | - Pietro Ferrara
- Unit of Pediatrics, Campus Bio-Medico University, Rome, Italy
| | - Lucilla Parnetti
- Section of Neurology, Laboratory of Experimental Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Costa
- Section of Neurology, Laboratory of Experimental Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
4
|
Li D, Zhang L, Tuo J, Zhang F, Tai Z, Liu X, Qiu X, Zhang H, Yang J, Wang J, Luo Z, Xu Z. PGC-1α Affects Epileptic Seizures by Regulating Mitochondrial Fusion in Epileptic Rats. Neurochem Res 2022; 48:1361-1369. [PMID: 36454394 DOI: 10.1007/s11064-022-03834-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/09/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), regulated by AMPK, is an important regulator of mitochondrial fusion. At present, whether the AMPK/PGC-1α signaling pathway regulates mitochondrial dynamics in epileptic rats is still unknown. METHODS Adult male Sprague-Dawley (SD) rats were randomly divided into fourgroups: the control group (0.9% saline, n = 5), the EP groups (lithium-pilocarpine was used to induce epilepsy, and tissues were harvested at 6 and 24 h, every time point, n = 5), the EP + Compound C group (the specific inhibitor of PGC-1α, 15 mg/kg in 2% DMSO, n = 5), and the EP + DMSO group (0.9% saline + 2% DMSO, n = 5). To investigate whether PGC-1α participates in seizures by regulating the expression of mitofusin1/2(MFN1/2)in rats. RESULTS In this study, the behavioral results indicate that the seizure susceptibility of the rats to epilepsy was increased when the expression of PGC-1α was inhibited. Subsequently, Western blot results suggested that the expression level of both MFN1 and MFN2 in the hippocampus was higher at 6 and 24 h after an epileptic seizure. Besides, the expression of PGC-1α and MFN2 was significantly decreased in the hippocampus when the epileptic rats were treated with Compound C. Furthermore, the immunofluorescence analysis of the localization of MFN1/2 and PGC-1α showed that MFN1/2 was mainly expressed in neurons but not astrocytes in the hippocampus and cerebral cortex of rats. Meanwhile, PGC-1α colocalized with the excitatory post-synaptic marker PSD95, suggesting that PGC-1α may regulate the seizure susceptibility of the rats by mediating excitatory post-synaptic signaling. CONCLUSION The AMPK/PGC-1α signaling pathway may play an important role in the lithium-pilocarpine-induced epileptic rat model by mediating the expression of fusion proteins.
Collapse
|
5
|
Li X, Wang Q, Zhang D, Wu D, Liu N, Chen T. Effects of long-term administration of Q808 on hippocampal transcriptome in healthy rats. Chem Pharm Bull (Tokyo) 2022; 70:642-649. [PMID: 35831127 DOI: 10.1248/cpb.c22-00357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epilepsy treatment with antiepileptic drugs (AEDs) is usually requires for many years. Q808 is an innovative antiepileptic chemical. It exerts effective antiepileptic effect against various epilepsy models. Exploring the gene transcriptomic profile of long-term treatment of Q808 is necessary. In the present study, hippocampus RNA-sequencing was performed to reveal the transcriptome profile of rats before and after treatment of Q808 for 28 days. Results confirmed 51 differentially expressed genes (DEGs) between Q808 and healthy control groups. Gene cluster analysis showed that most upregulated DEGs linked to response to drug and nucleus, most downregulated DEGs linked to locomotory, neuronal cell body, and drug binding. Most of DEGs were enriched in the signaling transduction, substance dependence, nervous system, and neurodegenerative disease pathways. Furthermore, quantitative real-time PCR analysis confirmed that Q808 significantly increased the expression of neuroprotective genes, such as Mdk, and decreased the mRNA levels of Penk, Drd1, and Adora2a, which are highly expressed in epilepsy models. In addition, Q808 decreased the mRNA expression of Pde10A and Drd2, which are known to be closely associated with schizophrenia. Our study may provide a theoretical basis to explore the effect of Q808 on the susceptibility to epilepsy and other neurological diseases.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University
| | - Qing Wang
- Jilin Provincial Academy of Traditional Chinese Medicine
| | - Dianwen Zhang
- Jilin Provincial Academy of Traditional Chinese Medicine
| | - Di Wu
- Jilin Provincial Academy of Traditional Chinese Medicine
| | - Ning Liu
- Jilin Provincial Academy of Traditional Chinese Medicine
| | - Tianli Chen
- School of Pharmacy, Changchun University of Chinese Medicine
| |
Collapse
|
6
|
Inhibition of PDE10A-Rescued TBI-Induced Neuroinflammation and Apoptosis through the cAMP/PKA/NLRP3 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3311250. [PMID: 35463083 PMCID: PMC9019408 DOI: 10.1155/2022/3311250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/22/2022] [Accepted: 03/22/2022] [Indexed: 11/18/2022]
Abstract
Phosphodiesterase 10A (PDE10A) is a dual-substrate phosphodiesterase that is highly expressed in the striatal complex. PDE10A is an important target for the treatment of ganglion dysfunction and neuroinflammation-related diseases, but its possible impact on traumatic brain injury (TBI) is still unclear. This study aims to investigate the protective effects of inhibiting PDE10A on neuroinflammation post-TBI injury and its possible molecular mechanism. The expression of PDE10A in rats and HT22 cells was determined by Western blotting. The neurological dysfunction of these rats was detected by Nissl staining, hematoxylin-eosin (HE) staining, and Morris water maze test. The activity of HT22 cells was measured by MTT. The findings of this study suggest that PDE10A is highly expressed in the brain tissue of TBI rats and HT22 cells induced by mechanical injury. Inhibition of PDE10A reduces the expression of interleukin-1β (IL-1β) and interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) in HT22 cells induced by mechanical injury to inhibit cell apoptosis. Simultaneously, inhibition of PDE10A in TBI rats reduces the time to find a visible platform in the same pool, while cAMP/PKA activator treatment alleviates all of the abovementioned phenomena. Additionally, it is further confirmed that inhibition of PDE10A activates the cAMP/PKA pathway and downregulates the expression of NRLP3. These findings demonstrate that inhibition of PDE10A exerts neuroprotection by inhibiting apoptosis and inflammation following TBI, at least partially by the cAMP/PKA/NLRP3 pathway.
Collapse
|
7
|
Ji X, Zeng Y, Wu J. The CB 2 Receptor as a Novel Therapeutic Target for Epilepsy Treatment. Int J Mol Sci 2021; 22:ijms22168961. [PMID: 34445666 PMCID: PMC8396521 DOI: 10.3390/ijms22168961] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 02/05/2023] Open
Abstract
Epilepsy is characterized by repeated spontaneous bursts of neuronal hyperactivity and high synchronization in the central nervous system. It seriously affects the quality of life of epileptic patients, and nearly 30% of individuals are refractory to treatment of antiseizure drugs. Therefore, there is an urgent need to develop new drugs to manage and control refractory epilepsy. Cannabinoid ligands, including selective cannabinoid receptor subtype (CB1 or CB2 receptor) ligands and non-selective cannabinoid (synthetic and endogenous) ligands, may serve as novel candidates for this need. Cannabinoid appears to regulate seizure activity in the brain through the activation of CB1 and CB2 cannabinoid receptors (CB1R and CB2R). An abundant series of cannabinoid analogues have been tested in various animal models, including the rat pilocarpine model of acquired epilepsy, a pentylenetetrazol model of myoclonic seizures in mice, and a penicillin-induced model of epileptiform activity in the rats. The accumulating lines of evidence show that cannabinoid ligands exhibit significant benefits to control seizure activity in different epileptic models. In this review, we summarize the relationship between brain CB2 receptors and seizures and emphasize the potential mechanisms of their therapeutic effects involving the influences of neurons, astrocytes, and microglia cells. The unique features of CB2Rs, such as lower expression levels under physiological conditions and high inducibility under epileptic conditions, make it an important target for future research on drug-resistant epilepsy.
Collapse
Affiliation(s)
- Xiaoyu Ji
- Brain Function and Disease Laboratory, Shantou University Medical College, Xin-Ling Road #22, Shantou 515041, China;
| | - Yang Zeng
- Medical Education Assessment and Research Center, Shantou University Medical College, Xin-Ling Road #22, Shantou 515041, China;
| | - Jie Wu
- Brain Function and Disease Laboratory, Shantou University Medical College, Xin-Ling Road #22, Shantou 515041, China;
- Correspondence: or
| |
Collapse
|
8
|
Kong Q, Zhang H, Wang M, Zhang J, Zhang Y. The TAAR1 inhibitor EPPTB suppresses neuronal excitability and seizure activity in mice. Brain Res Bull 2021; 171:142-149. [PMID: 33811954 DOI: 10.1016/j.brainresbull.2021.03.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
Epilepsy is a common neurological disease. G protein-coupled receptors (GPCRs) are extensively distributed and play an important role in human health by serving as therapeutic targets for various diseases. As one of the GPCRs, trace amine-associated receptor 1 (TAAR1) has recently aroused increasing interest as a potential therapeutic target for psychiatric disorders. However, the effect of TAAR1 on epileptic seizures remains unclear. We hypothesized that TAAR1 plays an important role in epilepsy and might represent a potential therapeutic target. In this study, we analyzed a mouse epilepsy model and patients with temporal lobe epilepsy (TLE) and observed substantially increased TAAR1 expression compared with the control group. In recordings of hippocampal slices, the TAAR1-specific inhibitor N-(3-ethoxyphenyl)-4-(pyrrolidin-1-yl)-3-(trifluoromethyl) benzamide (EPPTB) suppressed the excitability of hippocampal pyramidal neurons. EPPTB also reduced seizure-like events (SLEs) and seizure activity. Our results suggest that EPPTB attenuates seizure activity and that TAAR1 might be a potential drug target for individuals with epilepsy.
Collapse
Affiliation(s)
- Qingxia Kong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China; Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Hao Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Min Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China; Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Junchen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China.
| | - Yanke Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China; Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China.
| |
Collapse
|
9
|
Dong YY, Xia M, Wang L, Cui S, Li QB, Zhang JC, Meng SS, Zhang YK, Kong QX. Spatiotemporal Expression of SphK1 and S1PR2 in the Hippocampus of Pilocarpine Rat Model and the Epileptic Foci of Temporal Lobe Epilepsy. Front Cell Dev Biol 2020; 8:800. [PMID: 33134289 PMCID: PMC7578367 DOI: 10.3389/fcell.2020.00800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/28/2020] [Indexed: 01/03/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is a severe chronic neurological disease caused by abnormal discharge of neurons in the brain and seriously affect the long-term life quality of patients. Currently, new insights into the pathogenesis of TLE are urgently needed to provide more personalized and effective therapeutic strategies. Accumulating evidence suggests that sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate receptor 2 (S1PR2) signaling pathway plays a pivotal role in central nervous system (CNS) diseases. However, the precise altered expression of SphK1 and S1PR2 in TLE is remaining obscure. Here, we have confirmed the expression of SphK1 and S1PR2 in the pilocarpine-induced epileptic rat hippocampus and report for the first time the expression of SphK1 and S1PR2 in the temporal cortex of TLE patients. We found an increased expression of SphK1 in the brain from both epileptic rats and TLE patients. Conversely, S1PR2 expression level was markedly decreased. We further investigated the localization of SphK1 and S1PR2 in epileptic brains. Our study showed that both SphK1 and S1PR2 co-localized with activated astrocytes and neurons. Surprisingly, we observed different subcellular localization of SphK1 and S1PR2 in epileptic brain specimens. Taken together, our study suggests that the alteration of the SphK1/S1PR2 signaling axis is closely associated with the course of TLE and provides a new target for the treatment of TLE.
Collapse
Affiliation(s)
- Yuan-Yuan Dong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Min Xia
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lin Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Shuai Cui
- Department of Surgery, Weifang Medical University, Weifang, China
| | - Qiu-Bo Li
- Department of Pediatrics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Jun-Chen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Shu-Shu Meng
- Qingdao West Coast New Area Central Hospital, Qingdao, China
| | - Yan-Ke Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Qing-Xia Kong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
10
|
Krarup S, Mertz C, Jakobsen E, Lindholm SEH, Pinborg LH, Bak LK. Distinct effects on cAMP signaling of carbamazepine and its structural derivatives do not correlate with their clinical efficacy in epilepsy. Eur J Pharmacol 2020; 886:173413. [PMID: 32758572 DOI: 10.1016/j.ejphar.2020.173413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/07/2020] [Accepted: 07/23/2020] [Indexed: 01/06/2023]
Abstract
The antiepileptic sodium channel blocker, carbamazepine, has long been known to be able to attenuate cAMP signals. This could be of clinical importance since cAMP signaling has been shown to be involved in epileptogenesis and seizures. However, no information on the ability to affect cAMP signaling is available for the marketed structural derivatives, oxcarbazepine and eslicarbazepine acetate or their dominating metabolite, licarbazepine. Thus, we employed a HEK293 cell line stably expressing a cAMP biosensor to assess the effect of these two drugs on cAMP accumulation. We find that oxcarbazepine does not affect cAMP accumulation whereas eslicarbazepine acetate, surprisingly, is able to enhance cAMP accumulation. Since the transcription of ADCY8 (adenylyl cyclase isoform 8; AC8) has been found to be elevated in epileptic tissue from patients, we subsequently expressed AC8 in the HEK293 cells. In the AC8-expressing cells, oxcarbazepine was now able to attenuate whereas eslicarbazepine maintained its ability to increase cAMP accumulation. However, at all concentrations tested, licarbazepine demonstrated no effect on cAMP accumulation. Thus, we conclude that the effects exerted by carbamazepine and its derivatives on cAMP accumulation do not correlate with their clinical efficacy in epilepsy. However, this does not disqualify cAMP signaling per se as a potential disease-modifying drug target for epilepsy since more potent and selective inhibitors may be of therapeutic value.
Collapse
Affiliation(s)
- Sara Krarup
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Christoffer Mertz
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Emil Jakobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Sandy E H Lindholm
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Lars H Pinborg
- Epilepsy Clinic and Neurobiology Research Unit, Copenhagen University Hospital, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Lasse K Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark.
| |
Collapse
|
11
|
Lin Y, Ding Y, Jiang D, Li C, Huang X, Liu L, Xiao H, Vasudevan B, Chen Y. Genome-Wide Association of Genetic Variants With Refraction, Axial Length, and Corneal Curvature: A Longitudinal Study of Chinese Schoolchildren. Front Genet 2020; 11:276. [PMID: 32269590 PMCID: PMC7109285 DOI: 10.3389/fgene.2020.00276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/09/2020] [Indexed: 01/22/2023] Open
Abstract
Background Myopia is a common eye disorder that is approaching epidemic proportions worldwide. A genome-wide association study identified AREG (rs12511037), GABRR1 (rs13215566), and PDE10A (rs12206610) as being associated with refractive error in Asian populations. The present study investigated the associations between these three genetic variants and the occurrence and development of myopia, spherical equivalent refraction (SER), axial length (AL), and corneal curvature (CC) in a cohort of southeastern Chinese schoolchildren. Methods We examined and followed 550 children in grade 1 enrolled in the Wenzhou Epidemiology of Refractive Error (WERE) project. During the 4-year follow-up, non-cycloplegic refraction was evaluated twice each year, and the AL and CC were measured once every year. Age, sex, and the amounts of time spent on near work and outdoors were documented with a questionnaire. Sanger DNA sequencing was used to genotype single nucleotide polymorphisms (SNPs). SNPtest software was used to identify potential genetic variants associated with myopia, SER, AL, and CC. Ten thousand permutations were used to correct for multiple testing. Results In total, 469 children, including 249 (53.1%) boys and 220 (46.9%) girls, were included in analyses. The mean age of all the children was 6.33 ± 0.48 years. After adjusting for age, sex, time spent on near work and time spent outdoors, neither the genotypes nor the allele frequencies of the three SNPs were significantly associated with myopic shift, incident myopia or the change in SER. After adjusting for age, sex, near-work time and outdoor time with 10,000 permutations, the genotype AREG (rs12511037) was associated with an increase in AL (P′-values for the dominant, recessive, additive and general models were 0.0032, 0.0275, 0.0045, and 0.0099, respectively); the genotype PDE10A (rs12206610) was associated with a change in CC in the additive (P′ = 0.0096), dominant (P′ = 0.0096), and heterozygous models (P′ = 0.0096). Conclusion These findings preliminarily indicate that AREG SNP rs12511037 and PDE10A SNP rs12206610 are etiologically relevant for ocular traits, providing a basis for further exploration of the development of myopia and its molecular mechanism. However, elucidating the role of AREG and PDE10A in the pathogenesis of myopia requires further animal model and human genetic epidemiology studies. This trial is registered as ChiCTR1900020584 at www.Chictr.org.cn.
Collapse
Affiliation(s)
- Yaoyao Lin
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Yu Ding
- The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dandan Jiang
- The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chunchun Li
- The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoqiong Huang
- The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Linjie Liu
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Haishao Xiao
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | | | - Yanyan Chen
- The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Cyclic nucleotide phosphodiesterases: New targets in the metabolic syndrome? Pharmacol Ther 2020; 208:107475. [PMID: 31926200 DOI: 10.1016/j.pharmthera.2020.107475] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Metabolic diseases have a tremendous impact on human morbidity and mortality. Numerous targets regulating adenosine monophosphate kinase (AMPK) have been identified for treating the metabolic syndrome (MetS), and many compounds are being used or developed to increase AMPK activity. In parallel, the cyclic nucleotide phosphodiesterase families (PDEs) have emerged as new therapeutic targets in cardiovascular diseases, as well as in non-resolved pathologies. Since some PDE subfamilies inactivate cAMP into 5'-AMP, while the beneficial effects in MetS are related to 5'-AMP-dependent activation of AMPK, an analysis of the various controversial relationships between PDEs and AMPK in MetS appears interesting. The present review will describe the various PDE families, AMPK and molecular mechanisms in the MetS and discuss the PDEs/PDE modulators related to the tissues involved, thus supporting the discovery of original molecules and the design of new therapeutic approaches in MetS.
Collapse
|
13
|
Knopp C, Häusler M, Müller B, Damen R, Stoppe A, Mull M, Elbracht M, Kurth I, Begemann M. PDE10A mutation in two sisters with a hyperkinetic movement disorder - Response to levodopa. Parkinsonism Relat Disord 2019; 63:240-242. [PMID: 30777652 DOI: 10.1016/j.parkreldis.2019.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/12/2022]
Affiliation(s)
- C Knopp
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - M Häusler
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - B Müller
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - R Damen
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - A Stoppe
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - M Mull
- Department of Diagnostic and Interventional Neuroradiology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - M Elbracht
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - I Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - M Begemann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| |
Collapse
|