1
|
Nadi Moghadam N, Torkaman-Boutorabi A, Farhoudian A, Razaghi EM. Effects of acamprosate on alprazolam-induced conditioned place preference in male rats: The role of GABA and NMDA receptor subunits. Eur J Pharmacol 2025; 999:177643. [PMID: 40306538 DOI: 10.1016/j.ejphar.2025.177643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Alprazolam, a commonly prescribed benzodiazepine (BZD), poses a risk for abuse and has been linked to conditioned place preference (CPP). Research indicates that effective long-term treatments for alprazolam misuse are lacking. The mechanisms of tolerance and dependence for BZDs are similar to those seen with alcohol, involving gamma-aminobutyric acid (GABA) and glutamate neurotransmitter systems. Additionally, managing withdrawal symptoms and reducing relapse rates may be identical for both substances. Acamprosate's ability to reduce alcohol cravings and relapse has led this study to explore its potential as a treatment for the extinction and reinstatement of alprazolam-induced CPP. Accordingly, we evaluated the effects of different doses of acamprosate on the extinction period and reinstatement of alprazolam-induced CPP in male rats. We also assessed hippocampal gene expression of GABAA receptor (α1, α5, γ2) subunits and N-methyl-D-aspartate (NMDA) receptor (NR1, NR2A, NR2B) subunits after reinstatement, given alprazolam's action on these receptors. Alprazolam (1.5 mg/kg) could induce CPP in a 14-day paradigm. Acamprosate (20, 50, and 100 mg/kg) attenuated alprazolam-induced extinction period and reinstatement (P < 0.01). At the molecular level, acamprosate reduced the gene expression of α1 (P < 0.05) while increased α5 and γ2 subunits of GABAA receptors (p < 0.01). Besides, the gene expression of NR1, NR2A, and NR2B subunits of NMDA receptors were significantly enhanced by acamprosate (P < 0.001). These findings suggest that acamprosate is able to reduce the duration of extinction and reinstatement of alprazolam-induced CPP in male rats.
Collapse
Affiliation(s)
- Nasim Nadi Moghadam
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahita Torkaman-Boutorabi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ali Farhoudian
- Department of Psychiatry, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
2
|
Shih PC, Tzeng IS, Chen YC, Chen ML. Gastrodin Mitigates Ketamine-Induced Inhibition of F-Actin Remodeling and Cell Migration by Regulating the Rho Signaling Pathway. Biomedicines 2025; 13:649. [PMID: 40149625 PMCID: PMC11940296 DOI: 10.3390/biomedicines13030649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/16/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objects: Rho signaling plays a role in calcium-regulated cytoskeletal reorganization and cell movement, processes linked to neuronal function and cancer metastasis. Gastrodia elata, a traditional herbal medicine, can regulate glutamate-induced calcium influx in PC12 cells and influence cell function by modulating neuronal cytoskeleton remodeling via the monoaminergic system and Rho signaling. This study investigates the effects of gastrodin, a key component of Gastrodia elata, on Rho signaling, cytoskeleton remodeling, and cell migration in B35 and C6 cells. It also explores gastrodin's impact on Rho signaling in the prefrontal cortex of Sprague Dawley rats. Methods: B35 cells, C6 cells, and Sprague Dawley rats were treated with ketamine, gastrodin, or both. The expression of examined proteins from B35 cells, C6 cells, and the prefrontal cortex of Sprague Dawley rats were analyzed using immunoblotting. Immunofluorescent staining was applied to detect the phosphorylation of RhoGDI1. F-actin was stained using phalloidin-488 staining. Cell migration was analyzed using the Transwell and wound-healing assays. Results: Gastrodin reversed the ketamine-induced regulation of cell mobility inhibition, F-actin condensation, and Rho signaling modulation including Rho GDP dissociation inhibitor 1 (RhoGDI1); the Rho family protein (Ras homolog family member A (RhoA); cell division control protein 42 homolog (CDC42); Ras-related C3 botulinum toxin substrate 1(Rac1)); rho-associated, coiled-coil-containing protein kinase 1 (ROCK1); neural Wiskott-Aldrich syndrome protein (NWASP); myosin light chain 2 (MLC2); profilin1 (PFN1); and cofilin-1 (CFL1) in B35 and C6 cells. Similar modulations on Rho signaling were also observed in the prefrontal cortex of rats. Conclusions: Our findings show that gastrodin counteracts ketamine-induced disruptions in Rho signaling, cytoskeletal dynamics, and cell migration by regulating key components like RhoGDI1, ROCK1, MLC2, PFN1, and CFL1. This suggests the potential of gastrodin as a comprehensive regulator of cellular signaling.
Collapse
Affiliation(s)
- Ping-Cheng Shih
- Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
| | - Yi-Chyan Chen
- Department of Psychiatry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
| | - Mao-Liang Chen
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
| |
Collapse
|
3
|
Liu JR, Han XH, Yuki K, Soriano SG. Ketamine modulates disrupted in schizophrenia-1/glycogen synthase kinase-3β interaction. Front Mol Neurosci 2024; 17:1342233. [PMID: 38840775 PMCID: PMC11150584 DOI: 10.3389/fnmol.2024.1342233] [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: 11/21/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Disrupted in schizophrenia-1 (DISC1) is a scaffolding protein whose mutated form has been linked to schizophrenia, bipolar affective disorders, and recurrent major depression. DISC1 regulates multiple signaling pathways involved in neurite outgrowth and cortical development and binds directly to glycogen synthase kinase-3β (GSK-3β). Since ketamine activates GSK-3β, we examined the impact of ketamine on DISC1 and GSK-3β expression. Methods Postnatal day 7 rat pups were treated with ketamine with and without the non-specific GSK-3β antagonist, lithium. Cleaved-caspase-3, GSK-3β and DISC1 levels were measured by immunoblots and DISC1 co-localization in neurons by immunofluorescence. Binding of DISC1 to GSK-3β was determined by co-immunoprecipitation. Neurite outgrowth was determined by measuring dendrite and axon length in primary neuronal cell cultures treated with ketamine and lithium. Results Ketamine decreased DISC1 in a dose and time-dependent manner. This corresponded to decreases in phosphorylated GSK-3β, which implicates increased GSK-3β activity. Lithium significantly attenuated ketamine-induced decrease in DISC1 levels. Ketamine decreased co-immunoprecipitation of DISC1 with GSK-3β and axonal length. Conclusion These findings confirmed that acute administration of ketamine decreases in DISC1 levels and axonal growth. Lithium reversed this effect. This interaction provides a link between DISC1 and ketamine-induced neurodegeneration.
Collapse
Affiliation(s)
- Jia-Ren Liu
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Hui Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Koichi Yuki
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Anaesthesia, Harvard Medical School, Boston, MA, United States
| | - Sulpicio G. Soriano
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Anaesthesia, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Cavaleri D, Riboldi I, Crocamo C, Paglia G, Carrà G, Bartoli F. Evidence from preclinical and clinical metabolomics studies on the antidepressant effects of ketamine and esketamine. Neurosci Lett 2024; 831:137791. [PMID: 38670523 DOI: 10.1016/j.neulet.2024.137791] [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: 03/15/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
The antidepressant effects of ketamine and esketamine are well-documented. Nonetheless, most of the underlying molecular mechanisms have to be uncovered yet. In the last decade, metabolomics has emerged as a useful means to investigate the metabolic phenotype associated with depression as well as changes induced by antidepressant treatments. This mini-review aims at summarizing the main findings from preclinical and clinical studies that used metabolomics to investigate the metabolic effects of subanesthetic, antidepressant doses of ketamine and esketamine and their relationship with clinical response. Both animal and human studies report alterations in several metabolic pathways - including the tricarboxylic acid cycle, glycolysis, the pentose phosphate pathway, lipid metabolism, amino acid metabolism, the kynurenine pathway, and the urea cycle - following the administration of ketamine or its enantiomers. Although more research is needed to clarify commonalities and differences in molecular mechanisms of action between the racemic compound and its enantiomers, these findings comprehensively support an influence of ketamine and esketamine on mitochondrial and cellular energy production, membrane homeostasis, neurotransmission, and signaling. Metabolomics may thus represent a promising strategy to clarify molecular mechanisms underlying treatment-resistant depression and related markers of clinical response to ketamine and esketamine. This body of preclinical and clinical evidence, if further substantiated, has the potential to guide clinicians towards personalized approaches, contributing to new paradigms in the clinical management of depression.
Collapse
Affiliation(s)
- Daniele Cavaleri
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Ilaria Riboldi
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Cristina Crocamo
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Paglia
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Giuseppe Carrà
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy; Division of Psychiatry, University College London, 149 Tottenham Ct Rd, London W1T 7NF, United Kingdom
| | - Francesco Bartoli
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy.
| |
Collapse
|
5
|
Bahnamiri PJ, Hajizadeh Moghaddam A, Ranjbar M, Nazifi E. Effects of Nostoc commune extract on the cerebral oxidative and neuroinflammatory status in a mice model of schizophrenia. Biochem Biophys Rep 2024; 37:101594. [PMID: 38371525 PMCID: PMC10873873 DOI: 10.1016/j.bbrep.2023.101594] [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: 08/27/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 02/20/2024] Open
Abstract
Cyanobacterium Nostoc commune has long been used to alleviate various diseases. This research examines the effects of Nostoc commune extract (NCE) against behavioral disorders, cerebral oxidative stress, and inflammatory damage in the ketamine-induced schizophrenia model. Oral NCE administration (70 and 150 mg/kg/d) is performed after intraperitoneal ketamine injection (20 mg/kg) for 14 consecutive days. The forced swimming and open field tests are used to assess schizophrenia-like behaviors. After the behavioral test, dopamine (DA) level, oxidative stress markers, as well as the interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) expression are measured in the cerebral cortex. The results show that NCE treatment ameliorates KET-induced anxiety and depressive-like behaviors in OFT and FST, respectively. NCE considerably decreases the malondialdehyde (MDA) and DA levels and IL-6 and TNF-α expressions in mice with schizophrenia-like symptoms. Also, a significant increase is observed in the glutathione (GSH) level and catalase (CAT), superoxide dismutase (SOD), and glutathione reductase (GRx) activity in cerebral tissue. The present study shows that NCE treatment effectively improves KET-induced schizophrenia-like behaviors and oxidative and inflammatory damage. Therefore, NCE, via its bioactive constituents, could have strong neuroprotective effects in the schizophrenia-like model.
Collapse
Affiliation(s)
| | | | - Mojtaba Ranjbar
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Ehsan Nazifi
- Department of Plant Sciences, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
6
|
Zhan Q, Wang L, Liu N, Yuan Y, Deng L, Ding Y, Wang F, Zhou J, Xie L. Serum metabolomics study of narcolepsy type 1 based on ultra-performance liquid chromatography-tandem mass spectrometry. Amino Acids 2023; 55:1247-1259. [PMID: 37689600 PMCID: PMC10689557 DOI: 10.1007/s00726-023-03315-z] [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: 07/04/2023] [Accepted: 08/07/2023] [Indexed: 09/11/2023]
Abstract
Narcolepsy is a chronic and underrecognized sleep disorder characterized by excessive daytime sleepiness and cataplexy. Furthermore, narcolepsy type 1 (NT1) has serious negative impacts on an individual's health, society, and the economy. Currently, many sleep centers lack the means to measure orexin levels in the cerebrospinal fluid. We aimed to analyze the characteristics of metabolite changes in patients with NT1, measured by ultra-performance liquid chromatography-tandem mass spectrometry. A principal component analysis (PCA), an orthogonal partial least square discriminant analysis (OPLS-DA), t tests, and volcano plots were used to construct a model of abnormal metabolic pathways in narcolepsy. We identified molecular changes in serum specimens from narcolepsy patients and compared them with control groups, including dehydroepiandrosterone, epinephrine, N-methyl-D-aspartic acid, and other metabolites, based on an OPLS-loading plot analysis. Nine metabolites yielded an area under the receiver operating curve > 0.75. Meanwhile, seven abnormal metabolic pathways were correlated with differential metabolites, such as metabolic pathways; neuroactive ligand‒receptor interaction; and glycine, serine, and threonine metabolism. To our knowledge, this is the first study to reveal the characteristic metabolite changes in sera from NT1 patients for the selection of potential blood biomarkers and the elucidation of NT1 pathogenesis.
Collapse
Affiliation(s)
- Qingqing Zhan
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Lili Wang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Nan Liu
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yuqing Yuan
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Liying Deng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yongmin Ding
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Fen Wang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Jian Zhou
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Liang Xie
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
7
|
Maia MLF, Pantoja LVPS, Da Conceição BC, Machado-Ferraro KM, Gonçalves JKM, Dos Santos-Filho PM, Lima RR, Fontes-Junior EA, Maia CSF. Ketamine Clinical Use on the Pediatric Critically Ill Infant: A Global Bibliometric and Critical Review of Literature. J Clin Med 2023; 12:4643. [PMID: 37510758 PMCID: PMC10380297 DOI: 10.3390/jcm12144643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/14/2023] [Accepted: 05/29/2023] [Indexed: 07/30/2023] Open
Abstract
The developing central nervous system is vulnerable to several stimuli, especially psychotropic drugs. Sedation procedures during the developmental period are frequent in pediatric intensive care units (PICUs), in which the use of the sedative agent is still a challenge for the PICU team. Ketamine has been indicated for sedation in critically ill children with hemodynamic and ventilatory instabilities, but the possible neurobehavioral consequences related to this use are still uncertain. Here, we performed a bibliometric analysis with conventional metrics and a critical review of clinical findings to reveal a gap in the literature that deserves further investigation. We revealed that only 56 articles corresponded to the inclusion criteria of the study. The United States of America emerges as the main country within the scope of this review. In addition, professional clinical societies play a key role in the publications of scientific clinical findings through the specialist journals, which encourages the sharing of research work. The co-occurrence of keywords evidenced that the terms "sedation", "ketamine", and "pediatric" were the most frequent. Case series and review articles were the most prevalent study design. In the critical evaluation, the scarce studies highlight the need of use and post-use monitoring, which reinforces the importance of additional robust clinical studies to characterize the possible adverse effects resulting from ketamine anesthetic protocol in critically ill children.
Collapse
Affiliation(s)
- Mary Lucy Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| | - Lucas Villar Pedrosa Silva Pantoja
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| | - Brenda Costa Da Conceição
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| | - Kissila Márvia Machado-Ferraro
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| | - Jackeline Kerlice Mata Gonçalves
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| | - Paulo Monteiro Dos Santos-Filho
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Biological Science Institute, Federal University of Pará, Belém 66075-110, Pará, Brazil
| | - Enéas Andrade Fontes-Junior
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| | - Cristiane Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075-900, Pará, Brazil
| |
Collapse
|
8
|
Saito J, Zao H, Wu L, Iwasaki M, Sun Q, Hu C, Ishikawa M, Hirota K, Ma D. "Anti-cancer" effect of ketamine in comparison with MK801 on neuroglioma and lung cancer cells. Eur J Pharmacol 2023; 945:175580. [PMID: 36758782 DOI: 10.1016/j.ejphar.2023.175580] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Ketamine, a N-methyl-D-aspartate (NMDA) receptor antagonist, is commonly used to induce anaesthesia during cancer surgery and relieve neuropathic and cancer pain. This study was conducted to assess whether ketamine has any inhibiting effects on neuroglioma (H4) and lung cancer cells (A549) in vitro. The cultured H4 and A549 cells were treated with ketamine and MK801 (0.1, 1, 10, 100, or 1000 μM) for 24 h. The expressions of glutamate receptors on both types of cancer cells were assessed with qRT-PCR. In addition, cell proliferation and migration were assessed with cell counting Kit-8 and wound healing assays. Cyclin D1, matrix metalloproteinase 9 (MMP9), phosphorylation of extracellular signal-regulated kinase (pERK), and cleaved-caspase-3 expression together with reactive oxygen species (ROS) were also assessed with Western blot, immunostaining, and/or flowcytometry. NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors were expressed on both H4 and A549 cells. Ketamine inhibited cancer cell proliferation and migration in a dose-dependent manner by suppressing the cell cycle and inducing apoptosis. Ketamine decreased cyclin D1, pERK, and MMP9 expression. In addition, ketamine increased ROS and cleaved caspase-3 expression and induced apoptosis. The anti-cancer effect of ketamine was more pronounced in A549 cells when compared with H4 cells. MK801 showed similar effects to those of ketamine. Ketamine suppressed cell proliferation and migration in both neuroglioma and lung cancer cells, likely through the antagonization of NMDA receptors.
Collapse
Affiliation(s)
- Junichi Saito
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Hailin Zao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Lingzhi Wu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Masae Iwasaki
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Cong Hu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK.
| | - Masashi Ishikawa
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Kazuyoshi Hirota
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK; National Clinical Research Center for Child Health, Hangzhou, China.
| | | |
Collapse
|
9
|
Kim A, Gu SM, Lee H, Kim DE, Hong JT, Yun J, Cha HJ. Prenatal ketamine exposure impairs prepulse inhibition via arginine vasopressin receptor 1A-mediated GABAergic neuronal dysfunction in the striatum. Biomed Pharmacother 2023; 160:114318. [PMID: 36738499 DOI: 10.1016/j.biopha.2023.114318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Ketamine is a widely used anesthetic with N-methyl-D-aspartate (NMDA) receptor antagonism. Exposure to ketamine and NMDA receptor antagonists may induce psychosis. However, the mechanism underlying the effects of ketamine on the immature brain remains unclear. In this study, NMDA receptor antagonists, ketamine and methoxetamine, were administered to pregnant F344 rats (E17). These regimens induce psychosis-like behaviors in the offspring, such as hyperlocomotion induced by MK-801, a non-competitive NMDA receptor antagonist. We also observed that prepulse inhibition (PPI) was significantly reduced. Interestingly, ketamine administration increased the arginine vasopressin receptor 1A (Avpr1a) expression levels in the striatum of offspring with abnormal behaviors. Methoxetamine, another NMDA receptor antagonist, also showed similar results. In addition, we demonstrated a viral vector-induced Avpr1a overexpression in the striatum-inhibited PPI. In the striatum of offspring, ketamine or methoxetamine treatment increased glutamate decarboxylase 67 (GAD67) and δ-aminobutyric acid (GABA) levels. These results show that prenatal NMDA receptor antagonist treatment induces GABAergic neuronal dysfunction and abnormalities in sensorimotor gating via regulating Avpr1a expression in the striatum.
Collapse
Affiliation(s)
- Aeseul Kim
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Sun Mi Gu
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Haemiru Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Dong Eun Kim
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea.
| | - Hye Jin Cha
- College of Veterinary Medicine, Gyeongsang National University, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do 52828, the Republic of Korea.
| |
Collapse
|
10
|
LncRNA TUG1 Promoted Stabilization of BAG5 by Binding DDX3X to Exacerbate Ketamine-Induced Neurotoxicity. Neurotox Res 2022; 40:1989-2000. [PMID: 36151390 DOI: 10.1007/s12640-022-00580-w] [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: 05/30/2022] [Revised: 09/07/2022] [Accepted: 09/11/2022] [Indexed: 12/31/2022]
Abstract
As a clinically widely used anesthetic, ketamine (KET) has been reported to cause neurotoxicity in patients. Our work aimed to probe the function of long-chain non-coding RNA taurine-upregulated gene 1 (lncRNA TUG1) in KET-induced neurotoxicity. HT22 cells were subjected to KET to build the cell model. 3-(4, 5-Dimethylthiazolyl2)-2, 5-diphenyltetrazolium bromide (MTT) assay was employed to determine cell viability. Additionally, cell apoptosis was evaluated by flow cytometry. The binding relationships among TUG1, DEAD-box RNA helicase 3X (DDX3X), and Bcl-2-associated athanogene 5 (BAG5) were verified by RIP and RNA pull-down assays. Cell viability was impaired and cell apoptosis was increased in KET-treated HT22 cells accompanied by increased TUG1, DDX3X, and BAG5 expressions. TUG1 knockdown dramatically enhanced cell viability and repressed the of KET-induced apoptosis in HT22 cells, while TUG1 overexpression presented the opposite effects. In addition, we found that TUG1 promoted DDX3X expression via directly binding with DDX3X. As expected, DDX3X overexpression abolished the palliative effect of TUG1 knockdown on KET-induced neurotoxicity. Further research proved that TUG1 increased the stability of BAG5 through interacting with DDX3X. Finally, as expected, the moderating effect of TUG1 knockdown on KET-induced neuron injury was abolished by BAG5 overexpression. Taken together, TUG1 promoted BAG5 expression by binding DDX3X to exacerbate KET-induced neurotoxicity.
Collapse
|
11
|
Wang J, Liu Z. Research progress on molecular mechanisms of general anesthetic-induced neurotoxicity and cognitive impairment in the developing brain. Front Neurol 2022; 13:1065976. [PMID: 36504660 PMCID: PMC9729288 DOI: 10.3389/fneur.2022.1065976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
General anesthetics-induced neurotoxicity and cognitive impairment in developing brains have become one of the current research hotspots in the medical science community. The underlying mechanisms are complex and involve various related molecular signaling pathways, cell mediators, autophagy, and other pathological processes. However, few drugs can be directly used to treat neurotoxicity and cognitive impairment caused by general anesthetics in clinical practice. This article reviews the molecular mechanism of general anesthesia-induced neurotoxicity and cognitive impairment in the neonatal brain after surgery in the hope of providing critical references for the treatments of clinical diseases.
Collapse
Affiliation(s)
- Jiaojiao Wang
- Department of Anesthesiology, Baotou Central Hospital, Baotou, China,Baotou Clinical Medical College, Inner Mongolia Medical University, Baotou, China
| | - Zhihui Liu
- Department of Anesthesiology, Baotou Central Hospital, Baotou, China,*Correspondence: Zhihui Liu
| |
Collapse
|
12
|
Ferroptosis is involved in regulating perioperative neurocognitive disorders: emerging perspectives. J Neuroinflammation 2022; 19:219. [PMID: 36068571 PMCID: PMC9450301 DOI: 10.1186/s12974-022-02570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
Since the twenty-first century, the development of technological advances in anesthesia and surgery has brought benefits to human health. However, the adverse neurological effects of perioperative-related factors (e.g., surgical trauma, anesthesia, etc.) as stressors cannot be ignored as well. The nervous system appears to be more "fragile" and vulnerable to damage in developing and aging individuals. Ferroptosis is a novel form of programmed cell death proposed in 2012. In recent years, the regulation of ferroptosis to treat cancer, immune system disorders, and neurodegenerative diseases have seen an unprecedented surge of interest. The association of ferroptosis with perioperative neurocognitive disorders has also received much attention. Cognitive impairment can not only affect the individual's quality of life, but also impose a burden on the family and society. Therefore, the search for effective preventive and therapeutic methods to alleviate cognitive impairment caused by perioperative-related factors is a challenge that needs to be urgently addressed. In our review, we first briefly describe the connection between iron accumulation in neurons and impairment of brain function during development and aging. It is followed by a review of the pathways of ferroptosis, mainly including iron metabolism, amino acid metabolism, and lipid metabolism pathway. Furthermore, we analyze the connection between ferroptosis and perioperative-related factors. The surgery itself, general anesthetic drugs, and many other relevant factors in the perioperative period may affect neuronal iron homeostasis. Finally, we summarize the experimental evidence for ameliorating developmental and degenerative neurotoxicity by modulating ferroptosis. The suppression of ferroptosis seems to provide the possibility to prevent and improve perioperative neurocognitive impairment.
Collapse
|
13
|
Lisek M, Mackiewicz J, Sobolczyk M, Ferenc B, Guo F, Zylinska L, Boczek T. Early Developmental PMCA2b Expression Protects From Ketamine-Induced Apoptosis and GABA Impairments in Differentiating Hippocampal Progenitor Cells. Front Cell Neurosci 2022; 16:890827. [PMID: 35677757 PMCID: PMC9167922 DOI: 10.3389/fncel.2022.890827] [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: 03/06/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
PMCA2 is not expressed until the late embryonic state when the control of subtle Ca2+ fluxes becomes important for neuronal specialization. During this period, immature neurons are especially vulnerable to degenerative insults induced by the N-methyl-D-aspartate (NMDA) receptor blocker, ketamine. As H19-7 hippocampal progenitor cells isolated from E17 do not express the PMCA2 isoform, they constitute a valuable model for studying its role in neuronal development. In this study, we demonstrated that heterologous expression of PMCA2b enhanced the differentiation of H19-7 cells and protected from ketamine-induced death. PMCA2b did not affect resting [Ca2+]c in the presence or absence of ketamine and had no effect on the rate of Ca2+ clearance following membrane depolarization in the presence of the drug. The upregulation of endogenous PMCA1 demonstrated in response to PMCA2b expression as well as ketamine-induced PMCA4 depletion were indifferent to the rate of Ca2+ clearance in the presence of ketamine. Yet, co-expression of PMCA4b and PMCA2b was able to partially restore Ca2+ extrusion diminished by ketamine. The profiling of NMDA receptor expression showed upregulation of the NMDAR1 subunit in PMCA2b-expressing cells and increased co-immunoprecipitation of both proteins following ketamine treatment. Further microarray screening demonstrated a significant influence of PMCA2b on GABA signaling in differentiating progenitor cells, manifested by the unique regulation of several genes key to the GABAergic transmission. The overall activity of glutamate decarboxylase remained unchanged, but Ca2+-induced GABA release was inhibited in the presence of ketamine. Interestingly, PMCA2b expression was able to reverse this effect. The mechanism of GABA secretion normalization in the presence of ketamine may involve PMCA2b-mediated inhibition of GABA transaminase, thus shifting GABA utilization from energetic purposes to neurosecretion. In this study, we show for the first time that developmentally controlled PMCA expression may dictate the pattern of differentiation of hippocampal progenitor cells. Moreover, the appearance of PMCA2 early in development has long-standing consequences for GABA metabolism with yet an unpredictable influence on GABAergic neurotransmission during later stages of brain maturation. In contrast, the presence of PMCA2b seems to be protective for differentiating progenitor cells from ketamine-induced apoptotic death.
Collapse
Affiliation(s)
- Malwina Lisek
- Department of Molecular Neurochemistry, Medical University of Lodz, Łódz, Poland
| | - Joanna Mackiewicz
- Department of Molecular Neurochemistry, Medical University of Lodz, Łódz, Poland
| | - Marta Sobolczyk
- Department of Molecular Neurochemistry, Medical University of Lodz, Łódz, Poland
| | - Bozena Ferenc
- Department of Molecular Neurochemistry, Medical University of Lodz, Łódz, Poland
| | - Feng Guo
- Department of Pharmaceutical Toxicology, China Medical University, Shenyang, China
| | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, Łódz, Poland
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Łódz, Poland
- *Correspondence: Tomasz Boczek
| |
Collapse
|
14
|
Gale JR, Kosobucki GJ, Hartnett-Scott KA, Aizenman E. Imprecision in Precision Medicine: Differential Response of a Disease-Linked GluN2A Mutant to NMDA Channel Blockers. Front Pharmacol 2021; 12:773455. [PMID: 34776984 PMCID: PMC8581401 DOI: 10.3389/fphar.2021.773455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in N-methyl-d-aspartate receptors (NMDAR) subunits have been implicated in a growing number of human neurodevelopmental disorders. Previously, a de novo mutation in GRIN2A, encoding the GluN2A subunit, was identified in a patient with severe epilepsy and developmental delay. This missense mutation, which leads to GluN2A-P552R, produces significant dendrotoxicity in transfected rodent cortical neurons, as evidenced by pronounced dendritic blebbing. This injurious process can be prevented by treatment with the NMDA antagonist memantine. Given the increasing use of FDA approved NMDA antagonists to treat patients with GRIN mutations, who may have seizures refractory to traditional anti-epileptic drugs, we investigated whether additional NMDA antagonists were effective in attenuating neurotoxicity associated with GluN2A-P552R expression. Intriguingly, we found that while treatment with memantine can effectively block GluN2A-P552R-mediated dendrotoxicity, treatment with ketamine does not, despite the fact that both drugs work as open NMDAR channel blockers. Interestingly, we found that neurons expressing GluN2A-P552R were more vulnerable to an excitotoxic insult-an effect that, in this case, could be equally rescued by both memantine and ketamine. These findings suggest that GluN2A-P552R induced dendrotoxicity and increased vulnerability to excitotoxic stress are mediated through two distinct mechanisms. The differences between memantine and ketamine in halting GluN2A-P552R dendrotoxicity could not be explained by NMDA antagonist induced changes in MAP or Src kinase activation, previously shown to participate in NMDA-induced excitotoxicity. Our findings strongly suggest that not all NMDA antagonists may be of equal clinical utility in treating GRIN2A-mediated neurological disorders, despite a shared mechanism of action.
Collapse
Affiliation(s)
- Jenna R Gale
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Gabrielle J Kosobucki
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Karen A Hartnett-Scott
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Jin J, Wang R, Lin Q. The Increased Channel Activity of N-Methyl-D-Aspartate Receptors at Extrasynaptic Sites in the Anterior Cingulate Cortex of Neonatal Rats Following Prolonged Ketamine Exposure. J Pain Res 2021; 14:2381-2389. [PMID: 34393508 PMCID: PMC8360360 DOI: 10.2147/jpr.s320674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Ketamine is a dissociative anesthetic, commonly used for analgesia and anesthesia in a variety of pediatric procedures. It acts as a non-competitive antagonist to block ion channels of the N-methyl-D-aspartate receptors (NMDARs). Our previous study showed that repeated ketamine exposure developed a compensatory increase in NMDAR-mediated currents in neurons of the anterior cingulate cortex (ACC) of neonatal rats, and this increase was largely mediated by the GluN2B subunit-containing receptors, a predominant type of NMDARs during embryonic and early development of the brain. These data provide the molecular evidence to support that immature neurons are highly vulnerable to the development of apoptotic cell death after prolonged ketamine exposure. Methods Using whole-cell patch-clamp electrophysiology in an in vitro preparation of rat forebrain slices containing the ACC, the present study aimed at further determining whether GluN2B-containing NMDARs at extrasynaptic sites of immature neurons were the major target of ketamine for developing a compensatory increase in NMDAR-mediated synaptic transmission. Results Our major findings were that GluN2B subunits played a significant role in mediating ketamine-induced blockade of NMDAR-mediated currents in neonatal neurons and GluN2B-containing NMDARs expressed at extrasynaptic sites in neonatal neurons were the major player in compensatory enhancement of NMDAR-mediated currents after repeated ketamine exposure. Conclusion These results provide new evidence to strongly indicate that GluN2B-containing NMDARs at extrasynaptic sites are the key molecule contributing to the high vulnerability of the neonatal brain to ketamine-induced neurotoxic effects.
Collapse
Affiliation(s)
- Jianhui Jin
- Department of pain Management, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,Department of Psychology, The University of Texas at Arlington, Arlington, TX, USA
| | - Ruirui Wang
- Department of Psychology, The University of Texas at Arlington, Arlington, TX, USA
| | - Qing Lin
- Department of Psychology, The University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
16
|
Farkhondeh T, Samarghandian S, Roshanravan B, Peivasteh-Roudsari L. Impact of Curcumin on Traumatic Brain Injury and Involved Molecular Signaling Pathways. Recent Pat Food Nutr Agric 2021; 11:137-144. [PMID: 31288732 DOI: 10.2174/2212798410666190617161523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/14/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
Abstract
Traumatic Brain Injury (TBI) is one of the main causes of mortality and morbidity worldwide with no suitable treatment. The present study was designed to review the present literature about the protective effects of curcumin and the underlying mechanism against TBI. All published English language papers from beginning to 2019 were selected in this study. The findings indicate that curcumin may be effective against TBI outcomes by modulating the molecular signaling pathways involved in oxidative stress, inflammation, apoptosis, and autophagy. However, more experimental studies should be done to identify all mechanisms involved in the pathogenesis of TBI. Patents for Curcumin and chronic inflammation and traumatic brain injury management (WO2017097805A1 and US9101580B2) were published. In conclusion, the present study confirmed the potential therapeutic impact of curcumin for treating TBI.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Babak Roshanravan
- Medical Student, Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Leila Peivasteh-Roudsari
- Devision of Food Safety and Hygiene, Department of Environmental Health, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
The Relationship between the Concentration of Magnesium and the Presence of Depressive Symptoms and Selected Metabolic Disorders among Men over 50 Years of Age. Life (Basel) 2021; 11:life11030196. [PMID: 33802529 PMCID: PMC8001612 DOI: 10.3390/life11030196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 11/18/2022] Open
Abstract
Background: changes in the concentration of magnesium influence numerous processes in the body, such as hormone and lipid metabolism, nerve conduction, a number of biochemical pathways in the brain, and metabolic cycles. As a result, changes in magnesium concentration may contribute to the emergence of such pathologies as depressive and metabolic disorders, including hypertension, diabetes, and dyslipidemia. Methods: blood samples were taken from 342 men whose mean age was 61.66 ± 6.38 years. The concentrations of magnesium, lipid parameters, and glucose were determined using the spectrophotometric method. Anthropometric measurements were performed to determine each participant’s body mass index (BMI). Additionally, all participants completed two questionnaires: the Beck Depression Inventory and the author’s questionnaire. Results: abnormal levels of magnesium were found in 78 people. The analysis showed that these subjects more often suffered from metabolic disorders such as diabetes mellitus (p < 0.001), hypertension (p < 0.001), and depressive symptoms (p = 0.002) than participants with normal magnesium levels. Conclusion: our research showed that there is a relationship between abnormal levels of magnesium and the presence of self-reported conditions, such as diabetes, hypertension, and depressive symptoms among aging men. These findings may contribute to the improvement of the diagnosis and treatment of patients with these conditions.
Collapse
|
18
|
Culp C, Kim HK, Abdi S. Ketamine Use for Cancer and Chronic Pain Management. Front Pharmacol 2021; 11:599721. [PMID: 33708116 PMCID: PMC7941211 DOI: 10.3389/fphar.2020.599721] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022] Open
Abstract
Ketamine, an N-methyl-D-aspartate receptor antagonist, is widely known as a dissociative anesthetic and phencyclidine derivative. Due to an undesirable adverse event profile when used as an anesthetic it had widely fallen out of human use in favor of more modern agents. However, it has recently been explored for several other indications such as treatment resistant depression and chronic pain. Several recent studies and case reports compiled here show that ketamine is an effective analgesic in chronic pain conditions including cancer-related neuropathic pain. Of special interest is ketamine's opioid sparing ability by counteracting the central nervous system sensitization seen in opioid induced hyperalgesia. Furthermore, at the sub-anesthetic concentrations used for analgesia ketamine's safety and adverse event profiles are much improved. In this article, we review both the basic science and clinical evidence regarding ketamine's utility in chronic pain conditions as well as potential adverse events.
Collapse
Affiliation(s)
- Clayton Culp
- McGovern Medical School, University of Texas Health Science Center Houston (UTHealth), Houston, TX, United States
| | - Hee Kee Kim
- Division of Anesthesiology, Department of Pain Medicine, Critical Care and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Salahadin Abdi
- Division of Anesthesiology, Department of Pain Medicine, Critical Care and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
19
|
Xie L, Hu Y, Yan D, McQuillan P, Liu Y, Zhu S, Zhu Z, Jiang Y, Hu Z. The relationship between exposure to general anesthetic agents and the risk of developing an impulse control disorder. Pharmacol Res 2021; 165:105440. [PMID: 33493656 DOI: 10.1016/j.phrs.2021.105440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022]
Abstract
Most studies examining the effect of extended exposure to general anesthetic agents (GAAs) have demonstrated that extended exposure induces both structural and functional changes in the central nervous system. These changes are frequently accompanied by neurobehavioral changes that include impulse control disorders that are generally characterized by deficits in behavioral inhibition and executive function. In this review, we will.
Collapse
Affiliation(s)
- Linghua Xie
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuhan Hu
- Department of Cell Biology, Yale University, New Haven, CT, USA
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - P McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Centre, Penn State College of Medicine, Hershey, PA, USA
| | - Yue Liu
- Department of Anesthesiology, The Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhirui Zhu
- Department of Anesthesiology, The Children Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yilei Jiang
- Department of Anesthesiology, The Children Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
20
|
Wang R, Lin Q. Prolonged ketamine exposure induces enhanced excitatory GABAergic synaptic activity in the anterior cingulate cortex of neonatal rats. Neurosci Lett 2021; 745:135647. [PMID: 33444673 DOI: 10.1016/j.neulet.2021.135647] [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: 12/07/2020] [Accepted: 01/06/2021] [Indexed: 11/28/2022]
Abstract
Experimental studies have indicated that prolonged ketamine exposure in neonates at anesthetic doses causes neuronal apoptosis, which contributes to long-term impairments of learning and memory later in life. The neuronal excitotoxicity mediated by compensatory upregulation of N-methyl-d-aspartate receptors (NMDARs) is proposed to be the underlying mechanism. However, this view does not convincingly explain why excitotoxicity-related apoptotic injury develops selectively in immature neurons. We proposed that the GABAA receptors (GABAARs)-mediated excitatory synaptic signaling due to high expression of the Na+-K+-2Cl- co-transporter (NKCC1), occurring during the early neuronal development period, plays a distinct role in the susceptibility of immature neurons to ketamine-induced injury. Using whole-cell patch-clamp recordings from the forebrain slices containing the anterior cingulate cortex, we found that in vivo repeated ketamine administration significantly induced neuronal hyperexcitability in neonatal, but not adolescent, rats. Such hyperexcitability was accompanied by the increase both in GABAAR- and NMDAR-mediated synaptic transmissions. An interference with the NKCC1 by bumetanide treatment completely reversed these enhanced effects of ketamine exposure and blocked GABAAR-mediated postsynaptic current activity. Thus, these findings were significant as they showed, for the first time, that GABAAR-mediated excitatory action may contribute distinctly to neuronal excitotoxic effects of ketamine on immature neurons in the developing brain.
Collapse
Affiliation(s)
- Ruirui Wang
- Department of Psychology, The University of Texas at Arlington, TX, USA
| | - Qing Lin
- Department of Psychology, The University of Texas at Arlington, TX, USA.
| |
Collapse
|
21
|
Timic Stamenic T, Feseha S, Manzella FM, Wallace D, Wilkey D, Corrigan T, Fiedler H, Doerr P, Krishnan K, Raol YH, Covey DF, Jevtovic-Todorovic V, Todorovic SM. The T-type calcium channel isoform Ca v3.1 is a target for the hypnotic effect of the anaesthetic neurosteroid (3β,5β,17β)-3-hydroxyandrostane-17-carbonitrile. Br J Anaesth 2021; 126:245-255. [PMID: 32859366 PMCID: PMC7844375 DOI: 10.1016/j.bja.2020.07.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The mechanisms underlying the role of T-type calcium channels (T-channels) in thalamocortical excitability and oscillations in vivo during neurosteroid-induced hypnosis are largely unknown. METHODS We used patch-clamp electrophysiological recordings from acute brain slices ex vivo, recordings of local field potentials (LFPs) from the central medial thalamic nucleus in vivo, and wild-type (WT) and Cav3.1 knock-out mice to investigate the molecular mechanisms of hypnosis induced by the neurosteroid analogue (3β,5β,17β)-3-hydroxyandrostane-17-carbonitrile (3β-OH). RESULTS Patch-clamp recordings showed that 3β-OH inhibited isolated T-currents but had no effect on phasic or tonic γ-aminobutyric acid A currents. Also in acute brain slices, 3β-OH inhibited the spike firing mode more profoundly in WT than in Cav3.1 knockout mice. Furthermore, 3β-OH significantly hyperpolarised neurones, reduced the amplitudes of low threshold spikes, and diminished rebound burst firing only in WT mice. We found that 80 mg kg-1 i.p. injections of 3β-OH induced hypnosis in >60% of WT mice but failed to induce hypnosis in the majority of mutant mice. A subhypnotic dose of 3β-OH (20 mg kg-1 i.p.) accelerated induction of hypnosis by isoflurane only in WT mice, but had similar effects on the maintenance of isoflurane-induced hypnosis in both WT and Cav3.1 knockout mice. In vivo recordings of LFPs showed that a hypnotic dose of 3β-OH increased δ, θ, α, and β oscillations in WT mice in comparison with Cav3.1 knock-out mice. CONCLUSIONS The Cav3.1 T-channel isoform is critical for diminished thalamocortical excitability and oscillations that underlie neurosteroid-induced hypnosis.
Collapse
Affiliation(s)
- Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Simon Feseha
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Francesca M Manzella
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA; Neuroscience and Pharmacology Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Damon Wallace
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Davis Wilkey
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Timothy Corrigan
- Department of Pediatrics, Division of Neurology, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Hanna Fiedler
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Patricia Doerr
- Department of Anesthesiology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kathiresan Krishnan
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yogendra H Raol
- Department of Pediatrics, Division of Neurology, Translational Epilepsy Research Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA; Neuroscience and Pharmacology Graduate Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
22
|
Yao Y, Wang X, Gao J. LncRNA KCNQ1OT1 Sponges miR-206 to Ameliorate Neural Injury Induced by Anesthesia via Up-Regulating BDNF. Drug Des Devel Ther 2020; 14:4789-4800. [PMID: 33204065 PMCID: PMC7667201 DOI: 10.2147/dddt.s256319] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/23/2020] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Widely used in anesthesia, ketamine is reported to induce neurotoxicity in patients. This study aimed to investigate the molecular regulatory mechanism of long non-coding RNA (lncRNA) KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) in ameliorating ketamine-induced neural injury. MATERIALS AND METHODS Sprague-Dawley rats were intraperitoneally injected with ketamine to induce neuronal injury. PC-12 cells treated with ketamine were used as the cell model. Ketamine-induced aberrant expression of KCNQ1OT1, miR-206 and brain-derived neurotrophic factor (BDNF) were examined by quantitative real-time polymerase chain reaction (qRT-PCR). The effects of KCNQ1OT1 and miR-206 on ketamine-induced neural injury in PC-12 cells were then examined by MTT and LDH assay. The regulatory relationships between KCNQ1OT1 and miR-206, and miR-206 and BDNF were detected by dual-luciferase reporter assay. RESULTS Ketamine induced the apoptosis of neurons of the hippocampus in rats, and the apoptosis of PC-12 cells, accompanied by down-regulation of KCNQ1OT1 and BDNF expressions, and up-regulation of miR-206 expression. Overexpression of KCNQ1OT1 enhanced the resistance to apoptosis of PC-12 cells and significantly ameliorated ketamine-induced nerve injury, while transfection of miR-206 had opposite effects. Mechanistically, KCNQ1OT1 could target miR-206 and reduce its expression level, in turn indirectly increase the expression level of BDNF, and play a protective role in neural injury. CONCLUSION KCNQ1OT1/miR-206/BDNF axis is demonstrated to be an important regulatory mechanism in regulating ketamine-induced neural injury. Our study helps to clarify the mechanism by which ketamine exerts its toxicological effects and provides clues for the neuroprotection during anesthesia.
Collapse
Affiliation(s)
- Yao Yao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang441021, Hubei, People’s Republic of China
| | - Xuesong Wang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang441021, Hubei, People’s Republic of China
| | - Jin Gao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei College of Arts and Science, Xiangyang441021, Hubei, People’s Republic of China
| |
Collapse
|
23
|
Lisek M, Zylinska L, Boczek T. Ketamine and Calcium Signaling-A Crosstalk for Neuronal Physiology and Pathology. Int J Mol Sci 2020; 21:ijms21218410. [PMID: 33182497 PMCID: PMC7665128 DOI: 10.3390/ijms21218410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Ketamine is a non-competitive antagonist of NMDA (N-methyl-D-aspartate) receptor, which has been in clinical practice for over a half century. Despite recent data suggesting its harmful side effects, such as neuronal loss, synapse dysfunction or disturbed neural network formation, the drug is still applied in veterinary medicine and specialist anesthesia. Several lines of evidence indicate that structural and functional abnormalities in the nervous system caused by ketamine are crosslinked with the imbalanced activity of multiple Ca2+-regulated signaling pathways. Due to its ubiquitous nature, Ca2+ is also frequently located in the center of ketamine action, although the precise mechanisms underlying drug’s negative or therapeutic properties remain mysterious for the large part. This review seeks to delineate the relationship between ketamine-triggered imbalance in Ca2+ homeostasis and functional consequences for downstream processes regulating key aspects of neuronal function.
Collapse
|
24
|
Chen Q, Yan J, Xie W, Xie W, Li M, Ye Y. LncRNA LINC00641 Sponges miR-497-5p to Ameliorate Neural Injury Induced by Anesthesia via Up-Regulating BDNF. Front Mol Neurosci 2020; 13:95. [PMID: 32714145 PMCID: PMC7344214 DOI: 10.3389/fnmol.2020.00095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/06/2020] [Indexed: 12/31/2022] Open
Abstract
Introduction Ketamine, which is widely used in anesthesia, can induce cortical neurotoxicity in patients. This study aims to investigate the effects of long non-coding RNA LINC00641 on the ketamine-induced neural injury. Materials and Methods In this study, rat pheochromocytoma cells (PC12 cells) were used as a cell model and Sprague–Dawley postnatal day 7 rats were used for experiments in vivo. Ketamine-induced aberrant expression levels of LINC00641, miR-497-5p and brain-derived neurotrophic factor (BDNF) were examined by qRT-PCR. The effects of LINC00641 and miR-497-5p on ketamine-induced neural injury were then examined by MTT assays and TUNEL analysis. In addition, the activity of ROS and caspase-3 was measured. The regulatory relationships between LINC00641 and miR-497-5p, miR-497-5p and BDNF were detected by dual-luciferase reporter assay, respectively. Results Ketamine induced the apoptosis of PC12 cells, accompanied by down-regulation of LINC00641 and BDNF, and up-regulation of miR-497-5p. LINC00641 overexpression enhanced the resistance to the apoptosis of PC12 cells, while transfection of miR-497-5p had opposite effects. Furthermore, LINC00641 could bind to miR-497-5p and reduce its expression, but indirectly increase the BDNF expression, which was considered as a protective factor in neural injury and activated TrkB/PI3K/Akt pathway. Conclusion Collectively, LINC00641/miR-497-5p/BDNF axis was validated to be an important signaling pathway in modulating ketamine-induced neural injury.
Collapse
Affiliation(s)
- Qingxia Chen
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jingjia Yan
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wenji Xie
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wenqin Xie
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Meijun Li
- Department of Nursing, Quanzhou Medical College, Quanzhou, China
| | - Yanle Ye
- Department of Urology, The First Hospital of Quanzhou, Quanzhou, China
| |
Collapse
|
25
|
Ojeda J, Ávila A. Early Actions of Neurotransmitters During Cortex Development and Maturation of Reprogrammed Neurons. Front Synaptic Neurosci 2019; 11:33. [PMID: 31824293 PMCID: PMC6881277 DOI: 10.3389/fnsyn.2019.00033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
The development of the brain is shaped by a myriad of factors among which neurotransmitters play remarkable roles before and during the formation and maturation of synaptic circuits. Cellular processes such as neurogenesis, morphological development, synaptogenesis and maturation of synapses are temporary and spatially regulated by the local or distal influence of neurotransmitters in the developing cortex. Thus, research on this area has contributed to the understanding of fundamental mechanisms of brain development and to shed light on the etiology of various human neurodevelopmental disorders such as autism and Rett syndrome (RTT), among others. Recently, the field of neuroscience has been shaken by an explosive advance of experimental approaches linked to the use of induced pluripotent stem cells and reprogrammed neurons. This new technology has allowed researchers for the first time to model in the lab the unique events that take place during early human brain development and to explore the mechanisms that cause synaptopathies. In this context, the role of neurotransmitters during early stages of cortex development is beginning to be re-evaluated and a revision of the state of the art has become necessary in a time when new protocols are being worked out to differentiate stem cells into functional neurons. New perspectives on reconsidering the function of neurotransmitters include opportunities for methodological advances, a better understanding of the origin of mental disorders and the potential for development of new treatments.
Collapse
Affiliation(s)
- Jorge Ojeda
- Developmental Neurobiology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Ariel Ávila
- Developmental Neurobiology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| |
Collapse
|
26
|
Li Q, Qiu Z, Lu Y, Lu P, Wen J, Wang K, Zhao X, Li R, Zhang H, Zhang Y, Jia P, Fan P, Zhang Y, Zhang S, Lu H, Chen X, Liu Y, Zhang P. Edaravone protects primary-cultured rat cortical neurons from ketamine-induced apoptosis via reducing oxidative stress and activating PI3K/Akt signal pathway. Mol Cell Neurosci 2019; 100:103399. [DOI: 10.1016/j.mcn.2019.103399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 07/12/2019] [Accepted: 08/25/2019] [Indexed: 12/27/2022] Open
|
27
|
Antonov SA, Novosadova EV, Kobylyansky AG, Illarioshkin SN, Tarantul VZ, Grivennikov IA. Expression and Functional Properties of NMDA and GABA A Receptors during Differentiation of Human Induced Pluripotent Stem Cells into Ventral Mesencephalic Neurons. BIOCHEMISTRY (MOSCOW) 2019; 84:310-320. [PMID: 31221069 DOI: 10.1134/s0006297919030131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ionotropic glutamate and GABA receptors regulate the differentiation and determine the functional properties of mature neurons. Both insufficient and excessive activity of these neurotransmission systems are associated with various nervous system diseases. Our knowledge regarding the expression profiles of these receptors and the mechanisms of their regulation during the differentiation of specialized human neuron subtypes is limited. Here the expression profiles of the NMDA and GABAA receptor subunits were explored during in vitro differentiation of human induced pluripotent stem cells (iPSCs) into ventral mesencephalic neurons. The correlation between the neuronal maturation and the expression dynamics of these genes was investigated, and the functional activity of these receptors was assessed by calcium imaging. The role of NMDA and GABAA receptors in neurite outgrowth and the development of spontaneous activity was analyzed using the viral transduction of neural progenitors with the reporter genes TagGFP and TagRFP. The data indicate that agonists of the investigated receptors can be employed for optimization of existing protocols for neural differentiation of iPSCs, in particular for acceleration of neuronal maturation.
Collapse
Affiliation(s)
- S A Antonov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia.
| | - E V Novosadova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - A G Kobylyansky
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | | | - V Z Tarantul
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - I A Grivennikov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| |
Collapse
|
28
|
Robinson B, Gu Q, Ali SF, Dumas M, Kanungo J. Ketamine-induced attenuation of reactive oxygen species in zebrafish is prevented by acetyl l-carnitine in vivo. Neurosci Lett 2019; 706:36-42. [PMID: 31078678 DOI: 10.1016/j.neulet.2019.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 01/01/2023]
Abstract
Ketamine, an anesthetic, is a non-competitive antagonist of the calcium-permeable N-methyl-d-aspartate (NMDA) receptor. High concentrations of ketamine have been implicated in cardiotoxicity and neurotoxicity. Often, these toxicities are thought to be mediated by reactive oxygen species (ROS). However, findings to the contrary showing ketamine reducing ROS in mammalian cells and neurons in vitro, are emerging. Here, we determined the effects of ketamine on ROS levels in zebrafish larvae in vivo. Based on our earlier studies demonstrating reduction in ATP levels by ketamine, we hypothesized that as a calcium antagonist, ketamine would also prevent ROS generation, which is a by-product of ATP synthesis. To confirm that the detected ROS in a whole organism, such as the zebrafish larva, is specific, we used diphenyleneiodonium (DPI) that blocks ROS production by inhibiting the NADPH Oxidases (NOX). Upon 20 h exposure, DPI (5 and 10 μM) and ketamine at (1 and 2 mM) reduced ROS in the zebrafish larvae in vivo. Using acetyl l-carnitine (ALCAR), a dietary supplement, that induces mitochondrial ATP synthesis, we show elevated ROS generation with increasing ALCAR concentrations. Combined, ketamine and ALCAR counter-balanced ROS generation in the larvae suggesting that ketamine and ALCAR have opposing effects on mitochondrial metabolism, which may be key to maintaining ROS homeostasis in the larvae and affords ALCAR the ability to prevent ketamine toxicity. These results for the first time show ketamine's antioxidative and ALCAR's prooxidative effects in a live vertebrate.
Collapse
Affiliation(s)
- Bonnie Robinson
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Qiang Gu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Syed F Ali
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Melanie Dumas
- The Bionetics Corporation, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| |
Collapse
|
29
|
Ngwenya LB, Danzer SC. Impact of Traumatic Brain Injury on Neurogenesis. Front Neurosci 2019; 12:1014. [PMID: 30686980 PMCID: PMC6333744 DOI: 10.3389/fnins.2018.01014] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022] Open
Abstract
New neurons are generated in the hippocampal dentate gyrus from early development through adulthood. Progenitor cells and immature granule cells in the subgranular zone are responsive to changes in their environment; and indeed, a large body of research indicates that neuronal interactions and the dentate gyrus milieu regulates granule cell proliferation, maturation, and integration. Following traumatic brain injury (TBI), these interactions are dramatically altered. In addition to cell losses from injury and neurotransmitter dysfunction, patients often show electroencephalographic evidence of cortical spreading depolarizations and seizure activity after TBI. Furthermore, treatment for TBI often involves interventions that alter hippocampal function such as sedative medications, neuromodulating agents, and anti-epileptic drugs. Here, we review hippocampal changes after TBI and how they impact the coordinated process of granule cell adult neurogenesis. We also discuss clinical TBI treatments that have the potential to alter neurogenesis. A thorough understanding of the impact that TBI has on neurogenesis will ultimately be needed to begin to design novel therapeutics to promote recovery.
Collapse
Affiliation(s)
- Laura B Ngwenya
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, United States.,Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, United States.,Neurotrauma Center, University of Cincinnati Gardner Neuroscience Institute, Cincinnati, OH, United States
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Anesthesia, University of Cincinnati, Cincinnati, OH, United States.,Center for Pediatric Neuroscience, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
30
|
Robinson B, Dumas M, Gu Q, Kanungo J. N-acetylcysteine prevents ketamine-induced adverse effects on development, heart rate and monoaminergic neurons in zebrafish. Neurosci Lett 2018; 682:56-61. [PMID: 29890257 PMCID: PMC6102060 DOI: 10.1016/j.neulet.2018.06.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 01/11/2023]
Abstract
N-acetylcysteine, a precursor molecule of glutathione, is an antioxidant. Ketamine, a pediatric anesthetic, has been implicated in cardiotoxicity and neurotoxicity including modulation of monoaminergic systems in mammals and zebrafish. Here, we show that N-acetylcysteine prevents ketamine's adverse effects on development and monoaminergic neurons in zebrafish embryos. The effects of ketamine and N-acetylcysteine alone or in combination were measured on the heart rate, body length, brain serotonergic neurons and tyrosine hydroxylase-immunoreactive (TH-IR) neurons. In the absence of N-acetylcysteine, a concentration of ketamine that produces an internal embryo exposure level comparable to human anesthetic plasma concentrations significantly reduced heart rate and body length and those effects were prevented by N-acetylcysteine co-treatment. Ketamine also reduced the areas occupied by serotonergic neurons in the brain, whereas N-acetylcysteine co-exposure counteracted this effect. TH-IR neurons in the embryo brain and TH-IR cells in the trunk were significantly reduced with ketamine treatment, but not in the presence of N-acetylcysteine. In our continued search for compounds that can prevent ketamine toxicity, this study using specific endpoints of developmental toxicity, cardiotoxicity and neurotoxicity, demonstrates protective effects of N-acetylcysteine against ketamine's adverse effects. This is the first study that shows the protective effects of N-acetylcysteine on ketamine-induced developmental defects of monoaminergic neurons as observed in a whole organism.
Collapse
Affiliation(s)
- Bonnie Robinson
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Melanie Dumas
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Qiang Gu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| |
Collapse
|
31
|
Winlow W, Polese G, Moghadam HF, Ahmed IA, Di Cosmo A. Sense and Insensibility - An Appraisal of the Effects of Clinical Anesthetics on Gastropod and Cephalopod Molluscs as a Step to Improved Welfare of Cephalopods. Front Physiol 2018; 9:1147. [PMID: 30197598 PMCID: PMC6117391 DOI: 10.3389/fphys.2018.01147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/31/2018] [Indexed: 12/24/2022] Open
Abstract
Recent progress in animal welfare legislation stresses the need to treat cephalopod molluscs, such as Octopus vulgaris, humanely, to have regard for their wellbeing and to reduce their pain and suffering resulting from experimental procedures. Thus, appropriate measures for their sedation and analgesia are being introduced. Clinical anesthetics are renowned for their ability to produce unconsciousness in vertebrate species, but their exact mechanisms of action still elude investigators. In vertebrates it can prove difficult to specify the differences of response of particular neuron types given the multiplicity of neurons in the CNS. However, gastropod molluscs such as Aplysia, Lymnaea, or Helix, with their large uniquely identifiable nerve cells, make studies on the cellular, subcellular, network and behavioral actions of anesthetics much more feasible, particularly as identified cells may also be studied in culture, isolated from the rest of the nervous system. To date, the sorts of study outlined above have never been performed on cephalopods in the same way as on gastropods. However, criteria previously applied to gastropods and vertebrates have proved successful in developing a method for humanely anesthetizing Octopus with clinical doses of isoflurane, i.e., changes in respiratory rate, color pattern and withdrawal responses. However, in the long term, further refinements will be needed, including recordings from the CNS of intact animals in the presence of a variety of different anesthetic agents and their adjuvants. Clues as to their likely responsiveness to other appropriate anesthetic agents and muscle relaxants can be gained from background studies on gastropods such as Lymnaea, given their evolutionary history.
Collapse
Affiliation(s)
- William Winlow
- Department of Biology, University of Naples Federico II, Naples, Italy
- Institute of Ageing and Chronic Diseases, University of Liverpool, Liverpool, United Kingdom
- NPC Newton, Preston, United Kingdom
| | - Gianluca Polese
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Hadi-Fathi Moghadam
- Department of Physiology, Faculty of Medicine, Physiology Research Centre, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Anna Di Cosmo
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
32
|
Wang C, Han X, Liu F, Patterson TA, Hanig JP, Paule MG, Slikker W. Lipid profiling as an effective approach for identifying biomarkers/adverse events associated with pediatric anesthesia. Toxicol Appl Pharmacol 2018; 354:191-195. [PMID: 29550513 DOI: 10.1016/j.taap.2018.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/20/2018] [Accepted: 03/12/2018] [Indexed: 12/13/2022]
Abstract
Adverse effects related to central nervous system (CNS) function in pediatric populations may, at times, be difficult, if not impossible to evaluate. Prolonged anesthetic exposure affects brain excitability and anesthesia during the most sensitive developmental stages and has been associated with mitochondrial dysfunction, aberrant lipid metabolism and synaptogenesis, subsequent neuronal damage, as well as long-term behavioral deficits. There has been limited research evaluating whether and how anesthetic agents affect cellular lipids, the most abundant components of the brain other than water. Therefore, this review discusses: (1) whether the observed anesthetic-induced changes in lipid profiles seen in preclinical studies represents early signs of neurotoxicity; (2) the potential mechanisms underlying anesthetic-induced brain injury; and (3) whether lipid biomarker(s) identified in preclinical studies can serve as markers for the early clinical detection of anesthetic-induced neurotoxicity.
Collapse
Affiliation(s)
- Cheng Wang
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL 32827, USA
| | - Fang Liu
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079, USA
| | - Tucker A Patterson
- National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079, USA
| | - Joseph P Hanig
- Center for Drug Evaluation and Research/Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Merle G Paule
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079, USA
| | - William Slikker
- National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
33
|
Walters JL, Paule MG. Review of preclinical studies on pediatric general anesthesia-induced developmental neurotoxicity. Neurotoxicol Teratol 2017; 60:2-23. [DOI: 10.1016/j.ntt.2016.11.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/16/2016] [Accepted: 11/16/2016] [Indexed: 11/24/2022]
|
34
|
Lu J, Yao XQ, Luo X, Wang Y, Chung SK, Tang HX, Cheung CW, Wang XY, Meng C, Li Q. Monosialoganglioside 1 may alleviate neurotoxicity induced by propofol combined with remifentanil in neural stem cells. Neural Regen Res 2017; 12:945-952. [PMID: 28761428 PMCID: PMC5514870 DOI: 10.4103/1673-5374.208589] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Monosialoganglioside 1 (GM1) is the main ganglioside subtype and has neuroprotective properties in the central nervous system. In this study, we aimed to determine whether GM1 alleviates neurotoxicity induced by moderate and high concentrations of propofol combined with remifentanil in the immature central nervous system. Hippocampal neural stem cells were isolated from newborn Sprague-Dawley rats and treated with remifentanil (5, 10, 20 ng/mL) and propofol (1.0, 2.5, 5.0 μg/mL), and/or GM1 (12.5, 25, 50 μg/mL). GM1 reversed combined propofol and remifentanil-induced decreases in the percentage of 5-bromodeoxyuridine(+) cells and also reversed the increase in apoptotic cell percentage during neural stem cell proliferation and differentiation. However, GM1 with combined propofol and remifentanil did not affect β-tubulin(+) or glial fibrillary acidic protein(+) cell percentage during neural stem cell differentiation. In conclusion, we show that GM1 alleviates the damaging effects of propofol combined with remifentanil at moderate and high exposure concentrations in neural stem cells in vitro, and exerts protective effects on the immature central nervous system.
Collapse
Affiliation(s)
- Jiang Lu
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Xue-Qin Yao
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Xin Luo
- Department of Anesthesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.,Laboratory and Clinical Research Institute for Pain, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Sookja Kim Chung
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - He-Xin Tang
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Chi Wai Cheung
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.,Laboratory and Clinical Research Institute for Pain, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xian-Yu Wang
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Chen Meng
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Qing Li
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| |
Collapse
|
35
|
Cabrera OH, O'Connor SD, Swiney BS, Salinas-Contreras P, Manzella FM, Taylor GT, Noguchi KK. Caffeine combined with sedative/anesthetic drugs triggers widespread neuroapoptosis in a mouse model of prematurity. J Matern Fetal Neonatal Med 2016; 30:2734-2741. [PMID: 27924651 DOI: 10.1080/14767058.2016.1261400] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Caffeine (CAF) and sedative/anesthetic drugs (SADs) are often coadministered to premature infants in the neonatal intensive care unit (NICU). While SAD neurotoxicity in the developing brain is well established, it is not fully clear whether CAF interacts with SADs and whether this interaction is detrimental. Using a mouse model of prematurity, we hypothesized that CAF would increase apoptotic neurotoxicity when coadministered with SADs. METHODS Postnatal day 3 mice were treated with vehicle or 80 mg/kg CAF prior to challenge with 6 mg/kg midazolam, 40 mg/kg ketamine, or 40 μg/kg fentanyl. Six hours later, pups were sacrificed for activated caspase 3 (AC3) immunohistochemistry, and number of AC3 positive cells per mm3 throughout neocortex, hippocampus, caudate, thalamus, and colliculi was analyzed. RESULTS CAF caused a statistically significant increase in AC3 positive cells when coadministered with midazolam (p = 0.002), ketamine (p = 0.014), or fentanyl (p < 0.001). Our composite dataset suggests that the addition of CAF to these SADs has a supra-additive effect, causing more neurotoxicity than expected. CONCLUSIONS CAF may augment the neurotoxic action of SADs indicated for neonatal sedation/anesthesia in the NICU by triggering widespread apoptosis in the developing brains of premature infants.
Collapse
Affiliation(s)
- Omar Hoseá Cabrera
- a Department of Psychological Sciences , University of Missouri - St. Louis , St. Louis , MO , USA.,b Department of Psychiatry , Washington University in St. Louis School of Medicine , St. Louis , MO , USA
| | - Shawn David O'Connor
- c Edward Mallinckrodt Department of Pediatrics, Division of Newborn Medicine , Washington University in St. Louis School of Medicine and St. Louis Children's Hospital , St. Louis , MO , USA
| | - Brant Stephen Swiney
- b Department of Psychiatry , Washington University in St. Louis School of Medicine , St. Louis , MO , USA
| | - Patricia Salinas-Contreras
- b Department of Psychiatry , Washington University in St. Louis School of Medicine , St. Louis , MO , USA
| | - Francesca Maria Manzella
- a Department of Psychological Sciences , University of Missouri - St. Louis , St. Louis , MO , USA.,b Department of Psychiatry , Washington University in St. Louis School of Medicine , St. Louis , MO , USA
| | - George Townsend Taylor
- a Department of Psychological Sciences , University of Missouri - St. Louis , St. Louis , MO , USA
| | - Kevin Kiyoshi Noguchi
- b Department of Psychiatry , Washington University in St. Louis School of Medicine , St. Louis , MO , USA
| |
Collapse
|