1
|
Yang Y, Tuo J, Zhang J, Xu Z, Luo Z. Pathogenic genes implicated in sleep-related hypermotor epilepsy: a research progress update. Front Neurol 2024; 15:1416648. [PMID: 38966089 PMCID: PMC11222571 DOI: 10.3389/fneur.2024.1416648] [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: 04/12/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024] Open
Abstract
Sleep-related hypermotor epilepsy (SHE) is a focal epilepsy syndrome characterized by a variable age of onset and heterogeneous etiology. Current literature suggests a prevalence rate of approximately 1.8 per 100,000 persons. The discovery of additional pathogenic genes associated with SHE in recent years has significantly expanded the knowledge and understanding of its pathophysiological mechanisms. Identified SHE pathogenic genes include those related to neuronal ligand- and ion-gated channels (CHRNA4, CHRNB2, CHRNA2, GABRG2, and KCNT1), genes upstream of the mammalian target of rapamycin complex 1 signal transduction pathway (DEPDC5, NPRL2, NPRL3, TSC1, and TSC2), and other genes (CRH, CaBP4, STX1B, and PRIMA1). These genes encode proteins associated with ion channels, neurotransmitter receptors, cell signal transduction, and synaptic transmission. Mutations in these genes can result in the dysregulation of encoded cellular functional proteins and downstream neuronal dysfunction, ultimately leading to epileptic seizures. However, the associations between most genes and the SHE phenotype remain unclear. This article presents a literature review on the research progress of SHE-related pathogenic genes to contribute evidence to genotype-phenotype correlations in SHE and establish the necessary theoretical basis for future SHE treatments.
Collapse
Affiliation(s)
- Yufang Yang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jinmei Tuo
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhong Luo
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
2
|
Wang S, Cui D, Ling X, Hou Y, Sun J. Two novel variants of the STXBP1 and CHRNB2 genes identified in a Chinese boy with refractory seizures and developmental delay. Psychiatr Genet 2023; 33:206-212. [PMID: 37706497 DOI: 10.1097/ypg.0000000000000345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Autosomal dominant sleep-related hypermotor epilepsy is a rare disease caused by pathogenic variants of CHRNB2, CHRNA4, and CHRNA2 genes, with nocturnal frontal lobe epilepsy as the main symptoms. Syntaxin binding protein 1 (STXBP1) gene mutation can cause developmental and epileptic encephalopathy 4, mainly presenting as a developmental and epileptic encephalopathy. We performed the exome-targeted next-generation sequencing in our patient and identified two heterozygous variants: c.963 + 2T>C of STXBP1 and c.520_527delinsTGCTAC (p.R174Cfs*16) of CHRNB2. Molecular analysis was performed of the variant c.963 + 2T>C. Aberrantly spliced products were observed, proving the pathogenicity of this variant. Refractory seizures and developmental delay could be explained. Although the variant c.520_527delinsTGCTAC could cause the truncation of the proteins, it was ultimately determined to be nonpathogenic. The startle-like responses that occurred occasionally during the night were ultimately determined to be an uncommon phenotype caused by the STXBP1 variant.
Collapse
Affiliation(s)
- Sanmei Wang
- Department of Neurology and Development, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital
- Department of Neurology and Development, National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology
- Department of Neurology and Development, Beijing Key Laboratory of Pediatric Organ Failure
| | - Di Cui
- Beijing GrandOmics Biosciences Co., Ltd, Beijing, China
| | - Xiuxin Ling
- Beijing GrandOmics Biosciences Co., Ltd, Beijing, China
| | - Yu Hou
- Department of Neurology and Development, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital
- Department of Neurology and Development, National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology
- Department of Neurology and Development, Beijing Key Laboratory of Pediatric Organ Failure
| | - Jing Sun
- Department of Neurology and Development, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital
- Department of Neurology and Development, National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology
- Department of Neurology and Development, Beijing Key Laboratory of Pediatric Organ Failure
| |
Collapse
|
3
|
Hilscher MM, Mikulovic S, Perry S, Lundberg S, Kullander K. The alpha2 nicotinic acetylcholine receptor, a subunit with unique and selective expression in inhibitory interneurons associated with principal cells. Pharmacol Res 2023; 196:106895. [PMID: 37652281 DOI: 10.1016/j.phrs.2023.106895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) play crucial roles in various human disorders, with the α7, α4, α6, and α3-containing nAChR subtypes extensively studied in relation to conditions such as Alzheimer's disease, Parkinson's disease, nicotine dependence, mood disorders, and stress disorders. In contrast, the α2-nAChR subunit has received less attention due to its more restricted expression and the scarcity of specific agonists and antagonists for studying its function. Nevertheless, recent research has shed light on the unique expression pattern of the Chrna2 gene, which encodes the α2-nAChR subunit, and its involvement in distinct populations of inhibitory interneurons. This review highlights the structure, pharmacology, localization, function, and disease associations of α2-containing nAChRs and points to the unique expression pattern of the Chrna2 gene and its role in different inhibitory interneuron populations. These populations, including the oriens lacunosum moleculare (OLM) cells in the hippocampus, Martinotti cells in the neocortex, and Renshaw cells in the spinal cord, share common features and contribute to recurrent inhibitory microcircuits. Thus, the α2-nAChR subunit's unique expression pattern in specific interneuron populations and its role in recurrent inhibitory microcircuits highlight its importance in various physiological processes. Further research is necessary to uncover the comprehensive functionality of α2-containing nAChRs, delineate their specific contributions to neuronal circuits, and investigate their potential as therapeutic targets for related disorders.
Collapse
Affiliation(s)
- Markus M Hilscher
- Department of Immunology, Genetics and Pathology, Uppsala University, IGP/BMC, Box 815, 751 08 Uppsala, Sweden; Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Sanja Mikulovic
- Department of Immunology, Genetics and Pathology, Uppsala University, IGP/BMC, Box 815, 751 08 Uppsala, Sweden; Leibniz Institute for Neurobiology, Cognition & Emotion Laboratory, Magdeburg, Germany; German Center for Mental Health(DZPG), Germany
| | - Sharn Perry
- Department of Immunology, Genetics and Pathology, Uppsala University, IGP/BMC, Box 815, 751 08 Uppsala, Sweden; Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Australia
| | - Stina Lundberg
- Department of Immunology, Genetics and Pathology, Uppsala University, IGP/BMC, Box 815, 751 08 Uppsala, Sweden
| | - Klas Kullander
- Department of Immunology, Genetics and Pathology, Uppsala University, IGP/BMC, Box 815, 751 08 Uppsala, Sweden.
| |
Collapse
|
4
|
Bayanova M, Bolatov AK, Bazenova A, Nazarova L, Nauryzbayeva A, Tanko NM, Rakhimova S, Satvaldina N, Samatkyzy D, Kozhamkulov U, Kairov U, Akilzhanova A, Sarbassov D. Whole-Genome Sequencing Among Kazakhstani Children with Early-Onset Epilepsy Revealed New Gene Variants and Phenotypic Variability. Mol Neurobiol 2023; 60:4324-4335. [PMID: 37095367 PMCID: PMC10293429 DOI: 10.1007/s12035-023-03346-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023]
Abstract
In Kazakhstan, there is insufficient data on genetic epilepsy, which has its own clinical and management implications. Thus, this study aimed to use whole genome sequencing to identify and evaluate genetic variants and genetic structure of early onset epilepsy in the Kazakhstani pediatric population. In this study, for the first time in Kazakhstan, whole genome sequencing was carried out among epilepsy diagnosed children. The study involved 20 pediatric patients with early onset epilepsy and no established cause of the disease during the July-December, 2021. The average age at enrolment was 34.5 months, with a mean age at seizure onset of 6 months. Six patients (30%) were male, and 7 were familial cases. We identified pathogenic and likely pathogenic variants in 14 (70%) cases, among them, 6 novel disease gene variants (KCNQ2, CASK, WWOX, MT-CO3, GRIN2D, and SLC12A5). Other genes associated with the disease were SCN1A (x2), SLC2A1, ARX, CACNA1B, PCDH19, KCNT1, and CHRNA2. Identification of the genetic causes in 70% of cases confirms the general structure of the etiology of early onset epilepsy and the necessity of using NGS in diagnostics. Moreover, the study describes new genotype-phenotypic correlations in genetic epilepsy. Despite certain limitations of the study, it can be concluded that the genetic etiology of pediatric epilepsy in Kazakhstan is very broad and requires further research.
Collapse
Affiliation(s)
- Mirgul Bayanova
- University Medical Center CF, Kerey-Zhanibek Handar St. 5/1, Z05P3Y4, Astana, Kazakhstan
| | - Aidos K Bolatov
- University Medical Center CF, Kerey-Zhanibek Handar St. 5/1, Z05P3Y4, Astana, Kazakhstan.
- Astana Medical University, Beybitshilik St. 49A, Z10K9D9, Astana, Kazakhstan.
| | - Assiya Bazenova
- University Medical Center CF, Kerey-Zhanibek Handar St. 5/1, Z05P3Y4, Astana, Kazakhstan
| | - Lyazzat Nazarova
- University Medical Center CF, Kerey-Zhanibek Handar St. 5/1, Z05P3Y4, Astana, Kazakhstan
| | - Alissa Nauryzbayeva
- University Medical Center CF, Kerey-Zhanibek Handar St. 5/1, Z05P3Y4, Astana, Kazakhstan
| | - Naanlep Matthew Tanko
- University Medical Center CF, Kerey-Zhanibek Handar St. 5/1, Z05P3Y4, Astana, Kazakhstan
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan, 010000
| | - Saule Rakhimova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
| | - Nazerke Satvaldina
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
| | - Diana Samatkyzy
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
| | - Ulan Kozhamkulov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
| | - Ulykbek Kairov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
| | - Ainur Akilzhanova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
| | - Dos Sarbassov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
- School of Sciences and Humanities, Nazarbayev University, Kabanbay batyr Ave 53, Astana, Kazakhstan, 010000
| |
Collapse
|
5
|
Becchetti A, Grandi LC, Cerina M, Amadeo A. Nicotinic acetylcholine receptors and epilepsy. Pharmacol Res 2023; 189:106698. [PMID: 36796465 DOI: 10.1016/j.phrs.2023.106698] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/04/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Despite recent advances in understanding the causes of epilepsy, especially the genetic, comprehending the biological mechanisms that lead to the epileptic phenotype remains difficult. A paradigmatic case is constituted by the epilepsies caused by altered neuronal nicotinic acetylcholine receptors (nAChRs), which exert complex physiological functions in mature as well as developing brain. The ascending cholinergic projections exert potent control of forebrain excitability, and wide evidence implicates nAChR dysregulation as both cause and effect of epileptiform activity. First, tonic-clonic seizures are triggered by administration of high doses of nicotinic agonists, whereas non-convulsive doses have kindling effects. Second, sleep-related epilepsy can be caused by mutations on genes encoding nAChR subunits widely expressed in the forebrain (CHRNA4, CHRNB2, CHRNA2). Third, in animal models of acquired epilepsy, complex time-dependent alterations in cholinergic innervation are observed following repeated seizures. Heteromeric nAChRs are central players in epileptogenesis. Evidence is wide for autosomal dominant sleep-related hypermotor epilepsy (ADSHE). Studies of ADSHE-linked nAChR subunits in expression systems suggest that the epileptogenic process is promoted by overactive receptors. Investigation in animal models of ADSHE indicates that expression of mutant nAChRs can lead to lifelong hyperexcitability by altering i) the function of GABAergic populations in the mature neocortex and thalamus, ii) synaptic architecture during synaptogenesis. Understanding the balance of the epileptogenic effects in adult and developing networks is essential to plan rational therapy at different ages. Combining this knowledge with a deeper understanding of the functional and pharmacological properties of individual mutations will advance precision and personalized medicine in nAChR-dependent epilepsy.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, and NeuroMI (Milan Center of Neuroscience), University of Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy.
| | - Laura Clara Grandi
- Department of Biotechnology and Biosciences, and NeuroMI (Milan Center of Neuroscience), University of Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy.
| | - Marta Cerina
- Department of Biotechnology and Biosciences, and NeuroMI (Milan Center of Neuroscience), University of Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy.
| | - Alida Amadeo
- Department of Biosciences, University of Milano, Via Celoria 26, Milano 20133, Italy.
| |
Collapse
|
6
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
7
|
Meneghini S, Modena D, Colombo G, Coatti A, Milani N, Madaschi L, Amadeo A, Becchetti A. The β2V287L nicotinic subunit linked to sleep-related epilepsy differently affects fast-spiking and regular spiking somatostatin-expressing neurons in murine prefrontal cortex. Prog Neurobiol 2022; 214:102279. [DOI: 10.1016/j.pneurobio.2022.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 04/02/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022]
|
8
|
Abstract
The presence of unprovoked, recurrent seizures, particularly when drug resistant and associated with cognitive and behavioral deficits, warrants investigation for an underlying genetic cause. This article provides an overview of the major classes of genes associated with epilepsy phenotypes divided into functional categories along with the recommended work-up and therapeutic considerations. Gene discovery in epilepsy supports counseling and anticipatory guidance but also opens the door for precision medicine guiding therapy with a focus on those with disease-modifying effects.
Collapse
Affiliation(s)
- Luis A Martinez
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Drive, Houston, TX 77030, USA
| | - Yi-Chen Lai
- Department of Pediatrics, Section of Pediatric Critical Care Medicine, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Drive, Houston, TX 77030, USA
| | - J Lloyd Holder
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Drive, Houston, TX 77030, USA
| | - Anne E Anderson
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Drive, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Zheng JJ, Zhang TY, Liu HT, Huang ZX, Teng JM, Deng JX, Zhong JG, Qian X, Sheng XW, Ding JQ, He SQ, Zhao X, Ji WD, Qi DF, Li W, Zhang M. Cytisine Exerts an Anti-Epileptic Effect via α7nAChRs in a Rat Model of Temporal Lobe Epilepsy. Front Pharmacol 2021; 12:706225. [PMID: 34248648 PMCID: PMC8263902 DOI: 10.3389/fphar.2021.706225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Temporal lobe epilepsy (TLE) is a common chronic neurological disease that is often invulnerable to anti-epileptic drugs. Increasing data have demonstrated that acetylcholine (ACh) and cholinergic neurotransmission are involved in the pathophysiology of epilepsy. Cytisine, a full agonist of α7 nicotinic acetylcholine receptors (α7nAChRs) and a partial agonist of α4β2nAChRs, has been widely applied for smoking cessation and has shown neuroprotection in neurological diseases. However, whether cytisine plays a role in treating TLE has not yet been determined. Experimental Approach: In this study, cytisine was injected intraperitoneally into pilocarpine-induced epileptic rats for three weeks. Alpha-bungarotoxin (α-bgt), a specific α7nAChR antagonist, was used to evaluate the mechanism of action of cytisine. Rats were assayed for the occurrence of seizures and cognitive function by video surveillance and Morris water maze. Hippocampal injuries and synaptic structure were assessed by Nissl staining and Golgi staining. Furthermore, levels of glutamate, γ-aminobutyric acid (GABA), ACh, and α7nAChRs were measured. Results: Cytisine significantly reduced seizures and hippocampal damage while improving cognition and inhibiting synaptic remodeling in TLE rats. Additionally, cytisine decreased glutamate levels without altering GABA levels, and increased ACh levels and α7nAChR expression in the hippocampi of TLE rats. α-bgt antagonized the above-mentioned effects of cytisine treatment. Conclusion and Implications: Taken together, these findings indicate that cytisine exerted an anti-epileptic and neuroprotective effect in TLE rats via activation of α7nAChRs, which was associated with a decrease in glutamate levels, inhibition of synaptic remodeling, and improvement of cholinergic transmission in the hippocampus. Hence, our findings not only suggest that cytisine represents a promising anti-epileptic drug, but provides evidence of α7nAChRs as a novel therapeutic target for TLE.
Collapse
Affiliation(s)
- Jing-Jun Zheng
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Pharmacy, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
| | - Teng-Yue Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hong-Tao Liu
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ze-Xin Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jing-Mei Teng
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jing-Xian Deng
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jia-Gui Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xu Qian
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xin-Wen Sheng
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ji-Qiang Ding
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shu-Qiao He
- Department of Pharmacy, Maoming People's Hospital, Maoming, China
| | - Xin Zhao
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wei-Dong Ji
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - De-Feng Qi
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hop-ital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Wei Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Mei Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Abstract
OBJECTIVE Genetic variants of the neuronal nicotinic acetylcholine receptor (nAChR) cause autosomal dominant sleep-related hypermotor epilepsy. Approximately 30% of autosomal dominant sleep-related hypermotor epilepsy patients are medically intractable. In preclinical models, pathogenic nAChR variants cause a gain of function mutation with sensitivity to acetylcholine antagonists and agonists. Nicotine modifies the activity of nAChRs and can be used as targeted therapy. METHODS We reviewed next-generation sequencing epilepsy panels from a single laboratory (GeneDx) from patients at Children's Medical Center Dallas between 2011 and 2015 and identified patients with nAChR variants. Retrospective review of records included variant details, medical history, neuroimaging findings, and treatment history. RESULTS Twenty-one patients were identified. Four patients were prescribed nicotine patches for intractable seizures. Three of 4 patients had a clinical response, with >50% seizure reduction. CONCLUSIONS Treatment with a nicotine patch can be an effective therapy in epilepsy patients with nAChR gene variants. We propose consideration of transdermal nicotine treatment in intractable epilepsy with known nAChR variants as an experimental therapy. Further clinical trials are needed to fully define therapeutic effects.
Collapse
Affiliation(s)
- Jordana Fox
- Barrow Neurological Institute at 14524Phoenix Children's Hospital, Phoenix, AZ, USA
| | | | - Alison M Dolce
- 196285University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.,Children's Medical Center of Dallas, Dallas, TX, USA
| |
Collapse
|
11
|
Pavone P, Pappalardo XG, Ohazuruike UNN, Striano P, Parisi P, Corsello G, Marino SD, Ruggieri M, Parano E, Falsaperla R. Chromosome 15q BP4-BP5 Deletion in a Girl with Nocturnal Frontal Lobe Epilepsy, Migraine, Circumscribed Hypertrichosis, and Language Impairment. J Epilepsy Res 2020; 10:84-91. [PMID: 33659201 PMCID: PMC7903043 DOI: 10.14581/jer.20014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/06/2020] [Accepted: 11/25/2020] [Indexed: 01/01/2023] Open
Abstract
The 15q13.3 microdeletion (microdel15q13.3) syndrome (OMIM 612001) has been reported in healthy subjects as well as in individuals with a wide spectrum of clinical manifestations ranging from mild to severe neurological disorders, including developmental delay/intellectual disability, autism spectrum disorder, schizophrenia, epilepsy, behavioral problems and speech dysfunction. This study explored the link between this genomic rearrangement and nocturnal frontal lobe epilepsy (NFLE), which could improve the clinical interpretation. A clinical and genomic investigation was carried out on an 8-year-girl with a de novo deletion flanking the breakpoints (BPs) 4 and 5 of 15q13.3 detected by array comparative genomic hybridization analysis, affected by NFLE, migraine with aura, minor facial features, mild cognitive and language impairment, and circumscribed hypertrichosis. Literature survey of clinical studies was included. Nine years follow-up have displayed a benign course of the epileptic disorder with a progressive reduction and disappearance of the epileptic seizures, mild improvement of cognitive and language skills, partial cutaneous hypertrichosis regression, but stable ongoing of migraine episodes. A likely relationship between the BP4–BP5 deletion and NFLE with other symptoms presented by the girl is discussed together with a review of the literature on phenotypic features in microdel15q13.3.
Collapse
Affiliation(s)
- Piero Pavone
- Unit of Pediatrics and Pediatric Emergency, University Hospital "Policlinico-Vittorio Emanuele", Catania, Italy
| | - Xena Giada Pappalardo
- Unit of Catania, Institute for Biomedical Research and Innovation (IRIB), National Council of Research, Catania, Italy.,Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | | | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, IRCCS 'G. Gaslini' Institute, Genoa, Italy
| | - Pasquale Parisi
- Child Neurology, NESMOS Department, Faculty of Medicine & Psychology, "Sapienza" University, c/o Sant'Andrea Hospital, Rome, Italy
| | - Giovanni Corsello
- Department of Sciences for Health Promotion and Mother and Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | | | - Martino Ruggieri
- Unit of Pediatrics and Pediatric Emergency, University Hospital "Policlinico-Vittorio Emanuele", Catania, Italy
| | - Enrico Parano
- Unit of Catania, Institute for Biomedical Research and Innovation (IRIB), National Council of Research, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Neonatology University Hospital "Policlinico-Vittorio Emanuele", Catania, Italy
| |
Collapse
|
12
|
Nicotinic Receptors in Sleep-Related Hypermotor Epilepsy: Pathophysiology and Pharmacology. Brain Sci 2020; 10:brainsci10120907. [PMID: 33255633 PMCID: PMC7761363 DOI: 10.3390/brainsci10120907] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/19/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022] Open
Abstract
Sleep-related hypermotor epilepsy (SHE) is characterized by hyperkinetic focal seizures, mainly arising in the neocortex during non-rapid eye movements (NREM) sleep. The familial form is autosomal dominant SHE (ADSHE), which can be caused by mutations in genes encoding subunits of the neuronal nicotinic acetylcholine receptor (nAChR), Na+-gated K+ channels, as well as non-channel signaling proteins, such as components of the gap activity toward rags 1 (GATOR1) macromolecular complex. The causative genes may have different roles in developing and mature brains. Under this respect, nicotinic receptors are paradigmatic, as different pathophysiological roles are exerted by distinct nAChR subunits in adult and developing brains. The widest evidence concerns α4 and β2 subunits. These participate in heteromeric nAChRs that are major modulators of excitability in mature neocortical circuits as well as regulate postnatal synaptogenesis. However, growing evidence implicates mutant α2 subunits in ADSHE, which poses interpretive difficulties as very little is known about the function of α2-containing (α2*) nAChRs in the human brain. Planning rational therapy must consider that pharmacological treatment could have different effects on synaptic maturation and adult excitability. We discuss recent attempts towards precision medicine in the mature brain and possible approaches to target developmental stages. These issues have general relevance in epilepsy treatment, as the pathogenesis of genetic epilepsies is increasingly recognized to involve developmental alterations.
Collapse
|
13
|
Zupcic SG, Zupcic M, Duzel V, Simurina T, Sakic L, Grubjesic I, Tonković D, Udovic IS, Ferreri VM. The potential role of micro-RNA-211 in the pathogenesis of sleep-related hypermotor epilepsy. Med Hypotheses 2020; 143:110115. [DOI: https:/doi.org/10.1016/j.mehy.2020.110115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
|
14
|
Zhao D, Wang Q, Zhou WT, Wang LB, Yu H, Zhang KK, Chen LJ, Xie XL. PCB52 exposure alters the neurotransmission ligand-receptors in male offspring and contributes to sex-specific neurodevelopmental toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 264:114715. [PMID: 32402713 DOI: 10.1016/j.envpol.2020.114715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/09/2020] [Accepted: 04/30/2020] [Indexed: 06/11/2023]
Abstract
Polychlorinated biphenyls (PCBs) in the air are predominantly the less chlorinated congeners. Non-dioxin-like (NDL) low-chlorinated PCBs are more neurotoxic, and cause neurodevelopmental and neurobehavioral alterations in humans. However, the underlying mechanisms for this neurodevelopmental toxicity remain unknown. In the present study, Wistar rats were treated by gavage with PCB52 (1 mg/kg body weight) or corn oil from gestational day 7 to postnatal day 21. Both the body lengths and weights of the suckling rats at birth were significantly decreased by PCB52 treatment, suggesting developmental toxicity. Although no obvious histopathological changes were observed in the brain, using RNA-sequencing, 208 differentially expressed genes (DEGs) were identified in the striatum of PCB52-treated male offspring, while just 13 DEGs were identified in female offspring, suggesting sex-specific effects. Furthermore, using Gene Ontology enrichment analysis, neurodevelopmental processes, neurobehavioral alterations, and neurotransmission changes were enriched from the 208 DEGs in male offspring. Similarly, using Kyoto Encyclopedia of Genes and Genomes enrichment analysis, neuroactive ligand receptor interactions and multiple synapse pathways were enriched in male offspring, implying dysfunction of the neurotransmission system. Reductions in the protein expressions of these ligand receptors were also identified in the striatum, cerebral cortex, and hippocampus using western blotting methods. Taken together, our findings indicate that PCB52 exposure during gestation and lactation results in the abnormal expression of neurotransmission ligand-receptors in male offspring with a sex bias, and that this may contribute to neurodevelopmental toxicity.
Collapse
Affiliation(s)
- Dong Zhao
- Key Laboratory of Evidence Science (China University of Political Science and Law), Ministry of Education, Beijing, China
| | - Qi Wang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, No. 1838 North Guangzhou Road, 510515 Guangzhou, China
| | - Wen-Tao Zhou
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515 Guangzhou, China
| | - Li-Bin Wang
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515 Guangzhou, China
| | - Hao Yu
- The 2015 Class, 8-Year Program, The First Clinical Medical School, Southern Medical University, No. 1838 North Guangzhou Road, 510515 Guangzhou, China
| | - Kai-Kai Zhang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, No. 1838 North Guangzhou Road, 510515 Guangzhou, China
| | - Li-Jian Chen
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, No. 1838 North Guangzhou Road, 510515 Guangzhou, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515 Guangzhou, China.
| |
Collapse
|
15
|
Zupcic SG, Zupcic M, Duzel V, Simurina T, Sakic L, Grubjesic I, Tonković D, Udovic IS, Ferreri VM. The potential role of micro-RNA-211 in the pathogenesis of sleep-related hypermotor epilepsy. Med Hypotheses 2020; 143:110115. [PMID: 32763656 DOI: 10.1016/j.mehy.2020.110115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/12/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
Sleep-related hypermotor epilepsy (SHE) is a rare epileptic syndrome characterized by epileptic seizures which occur during the non-rapid eye movement (NREM) stage of sleep. It manifests with hypermotor semiology resembling violent limb movements and an asymmetric tonic-dystonic posture. The genes which are responsible for the autosomal dominant form of SHE (ADSHE) and whose function is to code the sub-unit of the neuronal acetylcholine receptor are well known. Considering that ADSHE is a prototype of SHE, it is thought that the dysfunction of the cortico-subcortical cholinergic network, which regulates the cycle of sleep, has a key role in the epileptogenesis of this syndrome. Namely, studies to date, have shown that the hypercholinergic activity is sufficient for the development of epileptic seizures, even though the exact mechanism remains to be elucidated. NREM parasomnias are sleep disorders that are the most difficult to differentiate from SHE due to a similar clinical presentation. Considering the clinical similarities, NREM occurrence and probable genetic connection, it is considered that fundamentally, both of these conditions share a common pathophysiological mechanism i.e. cholinergic dysfunction. The main difference between SHE and NREM parasomnias are the genuine epileptic seizures that are responsible for the semiology in SHE. These genuine seizures are not present in NREM parasomnias. Why this is so, remains to be elucidated. Considering that animal studies have shown that dynamic changes and the decreased levels of microRNA-211 contribute to epileptic seizures and to changes in cholinergic pathways, our hypothesis is that epileptic seizures and the development of epileptogenesis in SHE are a consequence of cholinergic dysfunction and decreased levels of microRNA-211 as opposed to NREM parasomnias where there is a stable level of microRNA-211, preventing epileptogenesis despite the cholinergic system dysfunction.
Collapse
Affiliation(s)
- Sandra Graf Zupcic
- Clinical Hospital Centre Rijeka, Clinic of Neurology, Rijeka, Croatia; University of Rijeka, Faculty of Medicine, Department of Physiology and Immunology, Rijeka, Croatia.
| | - Miroslav Zupcic
- University of Rijeka, Faculty of Medicine, Department of Physiology and Immunology, Rijeka, Croatia; Clinical Hospital Centre Rijeka, Clinic of Anesthesiology and Intensive Care Medicine, Rijeka, Croatia; J. J. Strossmayer University, Faculty of Medicine, Osijek, Croatia
| | - Viktor Duzel
- Barking, Havering and Redbridge University Hospitals NHS Trust, Department of Anaesthesia, London, United Kingdom
| | - Tatjana Simurina
- J. J. Strossmayer University, Faculty of Medicine, Osijek, Croatia; Department of Health Studies, University of Zadar, General Hospital Zadar, Department of Anesthesiology and Intensive Care Medicine, Zadar, Croatia
| | - Livija Sakic
- J. J. Strossmayer University, Faculty of Dental Medicine and Health, Osijek, Croatia; University Hospital "Sveti Duh", Clinic of Anesthesiology, Reanimatology and Intensive Care Medicine, Zagreb, Croatia
| | - Igor Grubjesic
- Clinical Hospital Centre Rijeka, Clinic of Anesthesiology and Intensive Care Medicine, Rijeka, Croatia
| | - Dinko Tonković
- School of Medicine, University of Zagreb, Croatia; Clinical Hospital Centre Zagreb, Clinic of Anesthesiology, Reanimatology and Intensive Care Medicine, Zagreb, Croatia
| | - Ingrid Sutic Udovic
- University of Rijeka, Faculty of Medicine, Department of Physiology and Immunology, Rijeka, Croatia
| | | |
Collapse
|
16
|
Germline Mutation Enrichment in Pathways Controlling Endothelial Cell Homeostasis in Patients with Brain Arteriovenous Malformation: Implication for Molecular Diagnosis. Int J Mol Sci 2020; 21:ijms21124321. [PMID: 32560555 PMCID: PMC7352422 DOI: 10.3390/ijms21124321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/19/2022] Open
Abstract
Brain arteriovenous malformation (bAVM) is a congenital defect affecting brain microvasculature, characterized by a direct shunt from arterioles to venules. Germline mutations in several genes related to transforming growth factor beta (TGF-β)/BMP signaling are linked to both sporadic and hereditary phenotypes. However, the low incidence of inherited cases makes the genetic bases of the disease unclear. To increase this knowledge, we performed a whole exome sequencing on five patients, on DNA purified by peripheral blood. Variants were filtered based on frequency and functional class. Those selected were validated by Sanger sequencing. Genes carrying selected variants were prioritized to relate these genes with those already known to be linked to bAVM development. Most of the prioritized genes showed a correlation with the TGF-βNotch signaling and vessel morphogenesis. However, two novel pathways related to cilia morphogenesis and ion homeostasis were enriched in mutated genes. These results suggest novel insights on sporadic bAVM onset and confirm its genetic heterogeneity. The high frequency of germline variants in genes related to TGF-β signaling allows us to hypothesize bAVM as a complex trait resulting from the co-existence of low-penetrance loci. Deeper knowledge on bAVM genetics can improve personalized diagnosis and can be helpful with genotype–phenotype correlations.
Collapse
|
17
|
Zhu L, Chen L, Xu P, Lu D, Dai S, Zhong L, Han Y, Zhang M, Xiao B, Chang L, Wu Q. Genetic and molecular basis of epilepsy-related cognitive dysfunction. Epilepsy Behav 2020; 104:106848. [PMID: 32028124 DOI: 10.1016/j.yebeh.2019.106848] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 02/02/2023]
Abstract
Epilepsy is a common neurological disease characterized by recurrent seizures. About 70 million people were affected by epilepsy or epileptic seizures. Epilepsy is a complicated complex or symptomatic syndromes induced by structural, functional, and genetic causes. Meanwhile, several comorbidities are accompanied by epileptic seizures. Cognitive dysfunction is a long-standing complication associated with epileptic seizures, which severely impairs quality of life. Although the definitive pathogenic mechanisms underlying epilepsy-related cognitive dysfunction remain unclear, accumulating evidence indicates that multiple risk factors are probably involved in the development and progression of cognitive dysfunction in patients with epilepsy. These factors include the underlying etiology, recurrent seizures or status epilepticus, structural damage that induced secondary epilepsy, genetic variants, and molecular alterations. In this review, we summarize several theories that may explain the genetic and molecular basis of epilepsy-related cognitive dysfunction.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Lu Chen
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Puying Xu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Di Lu
- Biomedicine Engineering Research Center, Kunming Medical University, 1168 Chun Rong West Road, Kunming, Yunnan 650500, PR China
| | - Shujuan Dai
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Lianmei Zhong
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Yanbing Han
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha, Hunan 410008, PR China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha, Hunan 410008, PR China
| | - Lvhua Chang
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China.
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China.
| |
Collapse
|
18
|
Cárdenas-Rodríguez N, Carmona-Aparicio L, Pérez-Lozano DL, Ortega-Cuellar D, Gómez-Manzo S, Ignacio-Mejía I. Genetic variations associated with pharmacoresistant epilepsy (Review). Mol Med Rep 2020; 21:1685-1701. [PMID: 32319641 PMCID: PMC7057824 DOI: 10.3892/mmr.2020.10999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/16/2020] [Indexed: 12/13/2022] Open
Abstract
Epilepsy is a common, serious neurological disorder worldwide. Although this disease can be successfully treated in most cases, not all patients respond favorably to medical treatments, which can lead to pharmacoresistant epilepsy. Drug-resistant epilepsy can be caused by a number of mechanisms that may involve environmental and genetic factors, as well as disease- and drug-related factors. In recent years, numerous studies have demonstrated that genetic variation is involved in the drug resistance of epilepsy, especially genetic variations found in drug resistance-related genes, including the voltage-dependent sodium and potassium channels genes, and the metabolizer of endogenous and xenobiotic substances genes. The present review aimed to highlight the genetic variants that are involved in the regulation of drug resistance in epilepsy; a comprehensive understanding of the role of genetic variation in drug resistance will help us develop improved strategies to regulate drug resistance efficiently and determine the pathophysiological processes that underlie this common human neurological disease.
Collapse
Affiliation(s)
- Noemí Cárdenas-Rodríguez
- Laboratory of Neuroscience, National Institute of Pediatrics, Ministry of Health, Coyoacán, Mexico City 04530, Mexico
| | - Liliana Carmona-Aparicio
- Laboratory of Neuroscience, National Institute of Pediatrics, Ministry of Health, Coyoacán, Mexico City 04530, Mexico
| | - Diana L Pérez-Lozano
- Laboratory of Neuroscience, National Institute of Pediatrics, Ministry of Health, Coyoacán, Mexico City 04530, Mexico
| | - Daniel Ortega-Cuellar
- Laboratory of Experimental Nutrition, National Institute of Pediatrics, Ministry of Health, Coyoacán, Mexico City 04530, Mexico
| | - Saúl Gómez-Manzo
- Laboratory of Genetic Biochemistry, National Institute of Pediatrics, Ministry of Health, Coyoacán, Mexico City 04530, Mexico
| | - Iván Ignacio-Mejía
- Laboratory of Translational Medicine, Military School of Health Graduates, Lomas de Sotelo, Militar, Mexico City 11200, Mexico
| |
Collapse
|