1
|
Agnorelli C, Spriggs M, Godfrey K, Sawicka G, Bohl B, Douglass H, Fagiolini A, Parastoo H, Carhart-Harris R, Nutt D, Erritzoe D. Neuroplasticity and psychedelics: A comprehensive examination of classic and non-classic compounds in pre and clinical models. Neurosci Biobehav Rev 2025; 172:106132. [PMID: 40185376 DOI: 10.1016/j.neubiorev.2025.106132] [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: 11/29/2024] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Neuroplasticity, the ability of the nervous system to adapt throughout an organism's lifespan, offers potential as both a biomarker and treatment target for neuropsychiatric conditions. Psychedelics, a burgeoning category of drugs, are increasingly prominent in psychiatric research, prompting inquiries into their mechanisms of action. Distinguishing themselves from traditional medications, psychedelics demonstrate rapid and enduring therapeutic effects after a single or few administrations, believed to stem from their neuroplasticity-enhancing properties. This review examines how classic psychedelics (e.g., LSD, psilocybin, N,N-DMT) and non-classic psychedelics (e.g., ketamine, MDMA) influence neuroplasticity. Drawing from preclinical and clinical studies, we explore the molecular, structural, and functional changes triggered by these agents. Animal studies suggest psychedelics induce heightened sensitivity of the nervous system to environmental stimuli (meta-plasticity), re-opening developmental windows for long-term structural changes (hyper-plasticity), with implications for mood and behavior. Translating these findings to humans faces challenges due to limitations in current imaging techniques. Nonetheless, promising new directions for human research are emerging, including the employment of novel positron-emission tomography (PET) radioligands, non-invasive brain stimulation methods, and multimodal approaches. By elucidating the interplay between psychedelics and neuroplasticity, this review informs the development of targeted interventions for neuropsychiatric disorders and advances understanding of psychedelics' therapeutic potential.
Collapse
Affiliation(s)
- Claudio Agnorelli
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK; Unit of Psychiatry, Department of Molecular and Developmental Medicine, University of Siena, Italy.
| | - Meg Spriggs
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Kate Godfrey
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Gabriela Sawicka
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Bettina Bohl
- Department of Bioengineering, Imperial College of London, UK
| | - Hannah Douglass
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Andrea Fagiolini
- Unit of Psychiatry, Department of Molecular and Developmental Medicine, University of Siena, Italy
| | | | - Robin Carhart-Harris
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK; Departments of Neurology and Psychiatry, Carhart-Harris Lab, University of California San Francisco, San Francisco, CA, USA
| | - David Nutt
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - David Erritzoe
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| |
Collapse
|
2
|
Wang P, Niu T, Huang D, Li Y, Jiang Z, Wang X, Liao L. Molecular mechanism of programmed cell death in drug-induced neuronal damage: A special focus on ketamine-induced neurotoxicity. Toxicology 2025; 513:154102. [PMID: 40015548 DOI: 10.1016/j.tox.2025.154102] [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: 01/01/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
In recent years, the abuse of ketamine as a recreational drug has been growing, and has become one of the most widely abused drugs. Continuous using ketamine poses a risk of drug addiction and complications such as attention deficit disorder, memory loss and cognitive decline. Ketamine-induced neurotoxicity is thought to play a key role in the development of these neurological complications. In this paper, we focus on the molecular mechanisms of ketamine-induced neurotoxicity. According to our analyses, drugs in causing neurotoxicity are closely associated with programmed cell death (PCD) such as apoptosis, autophagy, necroptosis, pyroptosis, and Ferroptosis. Therefore, this review will collate the existing mechanisms of programmed death in ketamine-induced neurotoxicity as well as explore the possible mechanisms by outlining the mechanisms of programmed death in other drug-induced neurotoxicity, which may be helpful in identifying potential therapeutic targets for neurotoxicity induced by ketamine abuse.
Collapse
Affiliation(s)
- Peipei Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Tong Niu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Degao Huang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yuanlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xia Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| | - Linchuan Liao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Yang TT, Guo ZW, Zhang F, Peng Y, Yu W, Gao GQ, Tian H, Zhang SJ, Liu JR. Lithium attenuates ketamine-induced long-term neurotoxicity through DISC1-mediated GSK-3β/β-catenin and ERK/CREB pathways. Toxicol Lett 2025; 406:50-62. [PMID: 40024338 DOI: 10.1016/j.toxlet.2025.02.012] [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: 08/21/2024] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Ketamine, an antagonist of N-methyl-D-aspartate receptor, is extensively employed in pediatric anesthesia. Multiple studies have shown that repeated ketamine exposure induces neuroapoptosis, synaptic changes and cognitive deficits during neurodevelopment. Therefore, it is essential to elucidate the mechanisms of ketamine-induced neurotoxicity and develop therapies to mitigate its harmful effects. Here, we investigated the role of disrupted in Schizophrenia 1 (DISC1) in ketamine-induced long-term neurotoxicity through a ketamine-exposed neuroapoptosis model. Neonatal rats received 2-5 intraperitoneal injections of ketamine (20 mg/kg b.w.) at 90 min intervals. Another cohort of pups received five intraperitoneal injections of ketamine (20 mg/kg×5 b.w.) with or without lithium (120 mg/kg×5 b.w.) at 90 min intervals over 6 h. Neuroapoptosis, DISC1-associated proteins expression in rats treated with ketamine, lithium, or a combination of both were detected, and the cognitive function of adolescent rats was evaluated by Morris water maze test. The length of dendrites and axons of primary neurons treated with lithium and ketamine were further measured. Results showed that ketamine time-dependently downregulated the levels of DISC1, pGSK-3β, β-catenin, pERK, pCREB and PSD95 in neonatal rats. Lithium could ameliorate neuroapoptosis, cognitive deficits and neurite growth inhibition triggered by ketamine. Mechanistically, lithium upregulated the levels of DISC1, PSD95 and GSK-3β/β-catenin and ERK/CREB signaling-related proteins. Consequently, lithium mitigated ketamine-induced long-term neurotoxicity by elevating DISC1 level and activating the GSK-3β/β-catenin and ERK/CREB signaling pathways.
Collapse
Affiliation(s)
- Ting-Ting Yang
- The Department of Clinical Laboratory, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China
| | - Zi-Wen Guo
- The Department of Clinical Laboratory, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China
| | - Fang Zhang
- The Center of Prenatal Diagnosis, Dongguan Maternal and Child Health Care Hospital, 99 Zhenxing Road, Zhushan District, Dongguan 523000, China
| | - Yu Peng
- The Department of Clinical Laboratory, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China
| | - Wei Yu
- The Department of Anesthesiology, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China
| | - Guang-Qiang Gao
- The Department of Clinical Laboratory, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China
| | - Hong Tian
- The Department of Clinical Laboratory, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China
| | - Shu-Jun Zhang
- The Department of Pathology, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China.
| | - Jia-Ren Liu
- The Department of Clinical Laboratory, the 4th Affiliated Hospital of Harbin Medical University, 32 Yi-Yuan Street, NanGang District, Harbin 150001, China.
| |
Collapse
|
4
|
Lima da Cruz RV, Leão RN, Moulin TC. Effects of psychedelics on neurogenesis and broader neuroplasticity: a systematic review. Mol Med 2024; 30:244. [PMID: 39701927 DOI: 10.1186/s10020-024-01013-4] [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: 07/17/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
In the mammalian brain, new neurons continue to be generated throughout life in a process known as adult neurogenesis. The role of adult-generated neurons has been broadly studied across laboratories, and mounting evidence suggests a strong link to the HPA axis and concomitant dysregulations in patients diagnosed with mood disorders. Psychedelic compounds, such as phenethylamines, tryptamines, cannabinoids, and a variety of ever-growing chemical categories, have emerged as therapeutic options for neuropsychiatric disorders, while numerous reports link their effects to increased adult neurogenesis. In this systematic review, we examine studies assessing neurogenesis or other neurogenesis-associated brain plasticity after psychedelic interventions and aim to provide a comprehensive picture of how this vast category of compounds regulates the generation of new neurons. We conducted a literature search on PubMed and Science Direct databases, considering all articles published until January 31, 2023, and selected articles containing both the words "neurogenesis" and "psychedelics". We analyzed experimental studies using either in vivo or in vitro models, employing classical or atypical psychedelics at all ontogenetic windows, as well as human studies referring to neurogenesis-associated plasticity. Our findings were divided into five main categories of psychedelics: CB1 agonists, NMDA antagonists, harmala alkaloids, tryptamines, and entactogens. We described the outcomes of neurogenesis assessments and investigated related results on the effects of psychedelics on brain plasticity and behavior within our sample. In summary, this review presents an extensive study into how different psychedelics may affect the birth of new neurons and other brain-related processes. Such knowledge may be valuable for future research on novel therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rafael V Lima da Cruz
- Neurodynamics Lab, Brain Institute (ICe), Universidade Federal do Rio Grande do Norte, Natal, Brazil.
| | - Richardson N Leão
- Neurodynamics Lab, Brain Institute (ICe), Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Thiago C Moulin
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
5
|
Huang H, Wang N, Lin JT, Qiu YK, Wu WF, Liu Q, Chen C, Wang HB, Liu YP, Dong W, Wan J, Zheng H, Zhou CH, Wu YQ. Repeated Ketamine Anesthesia during the Neonatal Period Impairs Hippocampal Neurogenesis and Long-Term Neurocognitive Function by Inhibiting Mfn2-Mediated Mitochondrial Fusion in Neural Stem Cells. Mol Neurobiol 2024; 61:5459-5480. [PMID: 38200350 DOI: 10.1007/s12035-024-03921-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
The mechanism of ketamine-induced neurotoxicity development remains elusive. Mitochondrial fusion/fission dynamics play a critical role in regulating neurogenesis. Therefore, this study was aimed to evaluate whether mitochondrial dynamics were involved in ketamine-induced impairment of neurogenesis in neonatal rats and long-term synaptic plasticity dysfunction. In the in vivo study, postnatal day 7 (PND-7) rats received intraperitoneal (i.p.) injection of 40 mg/kg ketamine for four consecutive times at 1 h intervals. The present findings revealed that ketamine induced mitochondrial fusion dysfunction in hippocampal neural stem cells (NSCs) by downregulating Mitofusin 2 (Mfn2) expression. In the in vitro study, ketamine treatment at 100 μM for 6 h significantly decreased the Mfn2 expression, and increased ROS generation, decreased mitochondrial membrane potential and ATP levels in cultured hippocampal NSCs. For the interventional study, lentivirus (LV) overexpressing Mfn2 (LV-Mfn2) or control LV vehicle was microinjected into the hippocampal dentate gyrus (DG) 4 days before ketamine administration. Targeted Mfn2 overexpression in the DG region could restore mitochondrial fusion in NSCs and reverse the inhibitory effect of ketamine on NSC proliferation and its faciliatory effect on neuronal differentiation. In addition, synaptic plasticity was evaluated by transmission electron microscopy, Golgi-Cox staining and long-term potentiation (LTP) recordings at 24 h after the end of the behavioral test. Preconditioning with LV-Mfn2 improved long-term cognitive dysfunction after repeated neonatal ketamine exposure by reversing the inhibitory effect of ketamine on synaptic plasticity in the hippocampal DG. The present findings demonstrated that Mfn2-mediated mitochondrial fusion dysfunction plays a critical role in the impairment of long-term neurocognitive function and synaptic plasticity caused by repeated neonatal ketamine exposure by interfering with hippocampal neurogenesis. Thus, Mfn2 might be a novel therapeutic target for the prevention of the developmental neurotoxicity of ketamine.
Collapse
Affiliation(s)
- He Huang
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Department of Anesthesiology and Perioperative Medicine, The First Affliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ning Wang
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jia-Tao Lin
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yong-Kang Qiu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wei-Feng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Chen Chen
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hai-Bi Wang
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yan-Ping Liu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wei Dong
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jie Wan
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Cheng-Hua Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
6
|
Mobinhosseini F, Salehirad M, Wallace Hayes A, Motaghinejad M, Hekmati M, Safari S, Gholami M. Curcumin-ZnO conjugated nanoparticles confer neuroprotection against ketamine-induced neurotoxicity. J Biochem Mol Toxicol 2024; 38:e23611. [PMID: 38084605 DOI: 10.1002/jbt.23611] [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: 01/02/2023] [Revised: 10/27/2023] [Accepted: 11/20/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Nanotechnology and its application to manipulate herbal compounds to design new neuroprotective agents to manage neurotoxicity has recently increased. Cur-ZnO conjugated nanoparticles were synthesized and used in an experimental model of ketamine-induced neurotoxicity. METHODS Cur-ZnO conjugated nanoparticles were chemically characterized, and the average crystalline size was determined. Forty-nine adult mice were divided into seven groups of seven animals each. Normal saline was given to control mice (group 1). Ketamine (25 mg/kg) was given to a second group. A third group of mice was given ketamine (25 mg/kg) in combination with curcumin (40 mg/kg), while mice in groups 4, 5, and 6 received ketamine (25 mg/kg) plus Cur-ZnO nanoparticles (10, 20, and 40 mg/kg). Group 7 received only ZnO (5 mg/kg). All doses were ip for 14 days. Hippocampal mitochondrial quadruple complex enzymes, oxidative stress, inflammation, and apoptotic characteristics were assessed. RESULTS Cur-ZnO nanoparticles and curcumin decreased lipid peroxidation, GSSG content, IL-1β, TNF-α, and Bax levels while increasing GSH and antioxidant enzymes like GPx, GR, and SOD while increasing Bcl-2 level and mitochondrial quadruple complex enzymes in ketamine treatment groups. CONCLUSION The neuroprotective properties of Cur-ZnO nanoparticles were efficient in preventing ketamine-induced neurotoxicity in the mouse brain. The nanoparticle form of curcumin (Cur-ZnO) required lower doses to produce neuroprotective effects against ketamine-induced toxicity than conventional curcumin.
Collapse
Affiliation(s)
- Fatemeh Mobinhosseini
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahsa Salehirad
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - A Wallace Hayes
- College of Public Health, University of South Florida, Tampa, Florida, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malak Hekmati
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Safari
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Gholami
- College of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
She YJ, Pan J, Peng LM, Ma L, Guo X, Lei DX, Wang HZ. Ketamine modulates neural stem cell differentiation by regulating TRPC3 expression through the GSK3β/β-catenin pathway. Neurotoxicology 2023; 94:1-10. [PMID: 36334642 DOI: 10.1016/j.neuro.2022.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Ketamine, a popular anesthetic, is often abused by people for its hallucinogenic effect. Thus, the safety of ketamine in pediatric populations has been called into question for potential neurotoxic effects. However, ketamine also has neuroprotective effects in many brain injury models. The differentiation of neural stem cells (NSCs) was influenced significantly by ketamine, but the molecular mechanism is still unclear. NSCs were extracted from the hippocampi of postnatal day 1 rats and treated with ketamine to induce NSCs differentiation. Our results found that ketamine promoted neuronal differentiation of NSCs dose-dependently in a small dose range (P < 0.001). The main types of neurons from NSCs were cholinergic (51 ± 4 %; 95 % CI: 41-61 %) and glutamatergic neurons (34 ± 3 %; 95 % CI: 27-42 %). Furthermore, we performed RNA sequencing to promise a more comprehensive understanding of the molecules regulated by ketamine. Finally, we combined bioimaging and multiple molecular biology techniques to clarify that ketamine influences NSC differentiation by regulating transient receptor potential canonical 3 (TRPC3) expressions. Ketamine dramatically repressed TRPC3 expression (MD [95 % CI]=0.67 [0.40-0.95], P < 0.001) with a significant increase of phosphorylated glycogen synthase kinase 3β (p-GSK3β; MD [95 % CI]=1.00 [0.74-1.27], P < 0.001) and a decrease of β-catenin protein expression (MD [95 % CI]=0.60 [0.32-0.89], P = 0.001), thereby promoting the differentiation of NSCs into neurons and inhibiting their differentiation into astrocytes. These results suggest that TRPC3 is necessary for ketamine to modulate NSC differentiation, which occurs partly via regulation of the GSK3β/β-catenin pathway.
Collapse
Affiliation(s)
- Ying-Jun She
- Department of Anesthesiology and Perioperative Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Junping Pan
- Department of Pharmacology, College of Basic Medicine, Jinan University, Guangzhou, China
| | - Liang-Ming Peng
- Department of Anesthesiology and Perioperative Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Ma
- Department of Cardiac Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Xinying Guo
- Department of Anesthesiology and Perioperative Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Dong-Xu Lei
- Department of Anesthesiology and Perioperative Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huai-Zhen Wang
- Department of Anesthesiology and Perioperative Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Vines L, Sotelo D, Johnson A, Dennis E, Manza P, Volkow ND, Wang GJ. Ketamine use disorder: preclinical, clinical, and neuroimaging evidence to support proposed mechanisms of actions. INTELLIGENT MEDICINE 2022; 2:61-68. [PMID: 35783539 PMCID: PMC9249268 DOI: 10.1016/j.imed.2022.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Ketamine, a noncompetitive NMDA receptor antagonist, has been exclusively used as an anesthetic in medicine and has led to new insights into the pathophysiology of neuropsychiatric disorders. Clinical studies have shown that low subanesthetic doses of ketamine produce antidepressant effects for individuals with depression. However, its use as a treatment for psychiatric disorders has been limited due to its reinforcing effects and high potential for diversion and misuse. Preclinical studies have focused on understanding the molecular mechanisms underlying ketamine's antidepressant effects, but a precise mechanism had yet to be elucidated. Here we review different hypotheses for ketamine's mechanism of action including the direct inhibition and disinhibition of NMDA receptors, AMPAR activation, and heightened activation of monoaminergic systems. The proposed mechanisms are not mutually exclusive, and their combined influence may exert the observed structural and functional neural impairments. Long term use of ketamine induces brain structural, functional impairments, and neurodevelopmental effects in both rodents and humans. Its misuse has increased rapidly in the past 20 years and is one of the most common addictive drugs used in Asia. The proposed mechanisms of action and supporting neuroimaging data allow for the development of tools to identify 'biotypes' of ketamine use disorder (KUD) using machine learning approaches, which could inform intervention and treatment.
Collapse
Affiliation(s)
| | | | - Allison Johnson
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Evan Dennis
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
9
|
Huang H, Zhao C, Hu Q, Liu Q, Sun YM, Chen C, Huang H, Zhou CH, Wu YQ. Neonatal Anesthesia by Ketamine in Neonatal Rats Inhibits the Proliferation and Differentiation of Hippocampal Neural Stem Cells and Decreases Neurocognitive Function in Adulthood via Inhibition of the Notch1 Signaling Pathway. Mol Neurobiol 2021; 58:6272-6289. [PMID: 34480336 DOI: 10.1007/s12035-021-02550-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 08/28/2021] [Indexed: 12/28/2022]
Abstract
The Notch signaling pathway plays an important role in the regulation of neurogenesis. The objective of this study was to investigate whether the Notch signaling pathway was involved in the neurogenesis impairment and long-term neurocognitive dysfunction caused by neonatal exposure to ketamine. On postnatal day 7 (PND-7), male Sprague-Dawley (SD) rats were intraperitoneally injected with 40 mg/kg ketamine four consecutive times (40 mg/kg × 4) at 1-h intervals. Notch ligand Jagged1 (0.5 mg/kg) and lentivirus overexpressing the Notch1 intracellular domain (LV-NICD1) were microinjected into the hippocampal dentate gyrus (DG) 1 h or 4 days before ketamine administration, respectively. The expression of Notch1 signaling pathway-related proteins was detected by Western blotting 24 h after ketamine administration. The proliferation and differentiation of the neural stem cells (NSCs) in the hippocampal DG were evaluated by double immunofluorescence staining 24 h after treatment. Moreover, changes in hippocampus-dependent spatial memory of 2-month-old rats were investigated with the Morris water maze test. Ketamine anesthesia in neonatal rats decreased the expression levels of Jagged1, Notch1, NICD1, and hairy enhancer of split 1 (Hes1); inhibited the proliferation and astrocytic differentiation of NSCs; and promoted the differentiation of neurons. Neonatal exposure to ketamine caused deficits in hippocampus-dependent spatial reference memory tasks in 2-month-old rats. Microinjection of Jagged1 or LV-NICD1 reversed the inhibitory effect of ketamine on the expression of Notch1-related proteins in the hippocampal DG, attenuated the ketamine-mediated decrease in NSC proliferation and differentiation, and improved the cognitive function of 2-month-old rats after neonatal exposure to ketamine. These results suggest that neonatal exposure to ketamine in rats inhibits the proliferation and differentiation of hippocampal NSCs and impairs neurocognitive function in adulthood. The Notch1 signaling pathway may be involved in the impairment of hippocampus-dependent learning and memory during adulthood caused by neonatal exposure to ketamine. These findings contribute to further understanding the neurotoxicity induced by neonatal exposure to ketamine and the underlying mechanisms.
Collapse
Affiliation(s)
- He Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Chao Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Qian Hu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yi-Man Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Chen Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Hui Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Cheng-Hua Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, People's Republic of China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, People's Republic of China.
| |
Collapse
|
10
|
Philippens IHCHM, Draaisma L, Baarends G, Krugers HJ, Vermetten E. Ketamine treatment upon memory retrieval reduces fear memory in marmoset monkeys. Eur Neuropsychopharmacol 2021; 50:1-11. [PMID: 33915317 DOI: 10.1016/j.euroneuro.2021.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022]
Abstract
Emotionally arousing experiences are retained very well as seen in posttraumatic stress disorder (PTSD). Various lines of evidence indicate that reactivation of these memories renders them labile which offers a potential time-window for intervention. We tested in non-human primates whether ketamine, administered during fear memory reactivation, affected passive (inhibitory) avoidance learning. For the consolidation of contextual emotional memory, the unescapable foot-shock paradigm in a passive avoidance task with two compartments (dark vs illuminated) was used. After entering the dark compartment, marmoset monkeys received four random foot-shocks (1 mA, 4 s) within 15-min. This stressful exposure increased the saliva cortisol and heart rate and impaired REM-sleep (p<0.05). One week later the monkeys were re-exposed to the stressful situation for the reconsolidation of the fearful experience. During the re-exposure the monkeys were treated with ketamine (0.5 mg/kg) or saline. In week 3, the monkeys were placed in the experimental setting to test their memory for the fearful experience. In contrast to the vehicle-treated monkeys, who avoided the dark compartment, the ketamine-treated monkeys entered the dark compartment that was previously associated with the fearful experience (p<0.05). Post-mortem analysis of the hippocampus showed that ketamine-treated animals exhibited less doublecortin positive neurons and BrdU-labeled cells in the dentate gyrus. This study reveals that a single low dose of ketamine, administered upon fear retrieval in monkeys, reduce contextual fear memory and attenuate neurogenesis in the hippocampus. These are important findings for considering ketamine as a potential candidate to target traumatic memories in PTSD.
Collapse
Affiliation(s)
- Ingrid H C H M Philippens
- Biomedical Primate Research Centre (BPRC), Animal Science Department, Lange Kleiweg 161, 2288 GJ Rijswijk, the Netherlands.
| | - Laurijn Draaisma
- Biomedical Primate Research Centre (BPRC), Animal Science Department, Lange Kleiweg 161, 2288 GJ Rijswijk, the Netherlands
| | - Guus Baarends
- Biomedical Primate Research Centre (BPRC), Animal Science Department, Lange Kleiweg 161, 2288 GJ Rijswijk, the Netherlands
| | - Harm J Krugers
- Faculty of Science, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| | - Eric Vermetten
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands; ARQ National Psychotrauma Center, Diemen, the Netherlands
| |
Collapse
|
11
|
Kalopita K, Armakolas A, Philippou A, Zarros A, Angelogianni P. Ketamine-induced neurotoxicity in neurodevelopment: A synopsis of main pathways based on recent in vivo experimental findings. J Anaesthesiol Clin Pharmacol 2021; 37:37-42. [PMID: 34103820 PMCID: PMC8174420 DOI: 10.4103/joacp.joacp_415_19] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022] Open
Abstract
Ketamine, a phencyclidine derivative and N-methyl-D-aspartate (NMDA) receptor antagonist, is widely used as an anesthetic, analgesic, and sedative agent in daily pediatric practice. Experimental studies have suggested that early prenatal or postnatal exposure to ketamine can induce neuroapoptosis, and establish neurobehavioral deficits that are evident in adulthood. However, most of the currently available clinical evidence is derived from retrospective and observational clinical studies. We, herein, attempt a brief review of the cellular and molecular mechanisms suggested to mediate ketamine-induced developmental neurotoxicity, utilizing a selected number of recent in vivo experimental evidence.
Collapse
Affiliation(s)
- Konstantina Kalopita
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Armakolas
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastassios Philippou
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Apostolos Zarros
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Panagoula Angelogianni
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Sha H, Peng P, Wei G, Wang J, Wu Y, Huang H. Neuroprotective Effects of Dexmedetomidine on the Ketamine-Induced Disruption of the Proliferation and Differentiation of Developing Neural Stem Cells in the Subventricular Zone. Front Pediatr 2021; 9:649284. [PMID: 34386466 PMCID: PMC8353121 DOI: 10.3389/fped.2021.649284] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 07/01/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Ketamine disrupts the proliferation and differentiation of developing neural stem cells (NSCs). Therefore, the safe use of ketamine in pediatric anesthesia has been an issue of increasing concern among anesthesiologists and children's parents. Dexmedetomidine (DEX) is widely used in sedation as an antianxiety agent and for analgesia. DEX has recently been shown to provide neuroprotection against anesthetic-induced neurotoxicity in the developing brain. The aim of this in vivo study was to investigate whether DEX exerted neuroprotective effects on the proliferation and differentiation of NSCs in the subventricular zone (SVZ) following neonatal ketamine exposure. Methods: Postnatal day 7 (PND-7) male Sprague-Dawley rats were equally divided into the following five groups: control group (n = 8), ketamine group (n = 8), 1 μg/kg DEX+ketamine group (n = 8), 5 μg/kg DEX+ketamine group (n = 8) and 10 μg/kg DEX+ketamine group (n = 8). Immediately after treatment, rats received a single intraperitoneal injection of BrdU, and the proliferation and differentiation of NSCs in the SVZ were assessed using immunostaining at 24 h after the BrdU injection. In the olfactory behavioral tests, rats in each group were raised until 2 months old, and the buried food test and olfactory memory test were performed. Results: The proliferation of NSCs and astrocytic differentiation in the SVZ were significantly inhibited at 24 h after repeated ketamine exposure in the neonatal period, and neuronal differentiation was markedly increased. Furthermore, pretreatment with moderately high (5 μg/kg) or high doses (10 μg/kg) of DEX reversed ketamine-induced disturbances in the proliferation and differentiation of NSCs. In the behavior tests, repeated neonatal ketamine exposure induced olfactory cognitive dysfunction in the adult stage, and moderately high and high doses of DEX reversed the olfactory cognitive dysfunction induced by ketamine. Conclusions: Based on the present findings, pretreatment with a moderately high (5 μg/kg) or high dose (10 μg/kg) of DEX may alleviate the developmental neurogenesis disorder in the SVZ at 24 h after repeated ketamine exposure and improve olfactory cognitive dysfunction in adulthood.
Collapse
Affiliation(s)
- Huanhuan Sha
- Department of Anesthesiology and Perioperative Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peipei Peng
- Department of Anesthesiology and Perioperative Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guohua Wei
- Department of Anesthesiology and Perioperative Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Wang
- Department of Anesthesiology and Perioperative Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - He Huang
- Department of Anesthesiology and Perioperative Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Yang J, Han W, Liu J, Yang C, Zhao WJ, Sun H, Pan YN, Chen HS, Cheng L, Jiang L. [Effect of advanced maternal age on development of hippocampal neural stem cells in offspring rats]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:1017-1026. [PMID: 32933637 PMCID: PMC7499440 DOI: 10.7499/j.issn.1008-8830.2003213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/05/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To study the effect of advanced maternal age (AMA) on the development of hippocampal neural stem cells in offspring rats. METHODS Ten 3-month-old and ten 12-month-old female Sprague-Dawley rats were housed individually with 3-month-old male rats (1:1, n=20), whose offspring rats were assigned to a control group and an AMA group. A total of 40 rats were randomly selected from each group. Immunofluorescence assay and Western blot were used to localize and determine the levels of protein expression of Nestin and doublecortin (DCX) on day 7 as well as neuronal nuclear antigen (NeuN) and glial fibrillary acidic protein (GFAP) on day 28 (n=8 for each marker). Immunofluorescence assay was also used to localize the hippocampal expression of polysialylated isoforms of neural cell adhesion molecule (PSA-NCAM) on day 14 (n=8 for each marker). RESULTS According to the Western blot results, the AMA group had significantly lower protein expression of DCX than the control group (P<0.05), while there were no significant differences in the protein expression of Nestin, NeuN, and GFAP between the two groups (P>0.05). According to the results of immunofluorescence assay, the AMA group had significantly lower protein expression of Nestin, DCX, and PSA-NCAM in the hippocampal dentate gyrus (DG) region than the control group (P<0.05), while there were no significant differences in the above indices in the hippocampal CA1 and CA3 regions between the two groups (P>0.05). The AMA group had significantly higher expression of NeuN in the hippocampal CA1 region than the control group (P<0.01), while there were no significant differences in the expression of NeuN in the hippocampal DG and CA3 regions between the two groups (P>0.05). The AMA group had significantly lower expression of GFAP in the hippocampal CA1, CA3, and DG regions than the control group (P<0.05). CONCLUSIONS AMA may cause inhibition of proliferation, survival, and migration of hippocampal neural stem cells. AMA may also affect their differentiation into neurons and astrocytes, which will eventually lead to developmental disorders of hippocampal neural stem cells in offspring rats.
Collapse
Affiliation(s)
- Jing Yang
- Department of Neurology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jia J, Zhu J, Yang Q, Wang Y, Zhang Z, Chen C. The Role of Histone Acetylation in the Sevoflurane-induced Inhibition of Neurogenesis in the Hippocampi of Young Mice. Neuroscience 2020; 432:73-83. [DOI: 10.1016/j.neuroscience.2020.02.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/27/2022]
|
15
|
Zhou X, Lv X, Zhang L, Yan J, Hu R, Sun Y, Xi S, Jiang H. Ketamine promotes the neural differentiation of mouse embryonic stem cells by activating mTOR. Mol Med Rep 2020; 21:2443-2451. [PMID: 32236601 PMCID: PMC7185302 DOI: 10.3892/mmr.2020.11043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 08/31/2018] [Indexed: 12/23/2022] Open
Abstract
Ketamine is a widely used general anesthetic and has been reported to demonstrate neurotoxicity and neuroprotection. Investigation into the regulatory mechanism of ketamine on influencing neural development is of importance for a better and safer way of relieving pain. Reverse transcription‑quantitative polymerase chain reaction and western blotting were used to detect the critical neural associated gene expression, and flow cytometry to detect the neural differentiation effect. Hence, in the present study the underlying mechanism of ketamine (50 nM) on neural differentiation of the mouse embryonic stem cell (mESC) line 46C was investigated. The results demonstrated that a low dose of ketamine (50 nM) promoted the differentiation of mESCs to neural stem cells (NSCs) and activated mammalian target of rapamycin (mTOR) by upregulating the expression levels of phosphorylated (p)‑mTOR. Furthermore, inhibition of the mTOR signaling pathway by rapamycin or knockdown of mTOR suppressed neural differentiation. A rescue experiment further confirmed that downregulation of mTOR inhibited the promotion of neural differentiation induced by ketamine. Taken together, the present study indicated that a low level of ketamine upregulated p‑mTOR expression levels, promoting neural differentiation.
Collapse
Affiliation(s)
- Xuhui Zhou
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| | - Xiang Lv
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| | - Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| | - Rong Hu
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| | - Siwei Xi
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, P.R. China
| |
Collapse
|
16
|
Xie R, Hong S, Ye Y, Wang X, Chen F, Yang L, Yan Y, Liao L. Ketamine Affects the Expression of ErbB4 in the Hippocampus and Prefrontal Cortex of Rats. J Mol Neurosci 2020; 70:962-967. [PMID: 32096126 DOI: 10.1007/s12031-020-01502-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 12/16/2022]
Abstract
Schizophrenia is a severe chronic neuropsychiatric disorder, and its exact pathogenesis remains unclear. This study investigated the effect of ketamine on the expression of ErbB4 (considered a schizophrenia candidate gene) in the hippocampus and prefrontal cortex of rats. Rats were randomly divided into four groups: control, low-dose, medium-dose and high-dose groups. The low-dose, medium-dose and high-dose groups were intraperitoneally injected with 15 mg/kg, 30 mg/kg and 60 mg/kg ketamine, respectively, twice a day (9:00 a.m. and 9:00 p.m.); the control group was administered normal saline. The treatment lasted 7 days. After treatment, rats were euthanized, and their brain tissues were collected and then analyzed by immunohistochemistry. The results of immunohistochemistry staining demonstrated that the ErbB4 protein was expressed exclusively in the CA3 region of the hippocampus and the Cg1 region of the prefrontal cortex. Ketamine administration significantly decreased the expression of ErbB4 in a dose-dependent manner. The high-dose ketamine treatment was found to be optimal for establishing a rat model for schizophrenia. Ketamine induced symptoms similar to schizophrenia in humans. The ketamine-induced rat model for schizophrenia constructed in this study provides novel insights to better understand the pathogenic mechanisms of schizophrenia and aid in drug discovery.
Collapse
Affiliation(s)
- Runfang Xie
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, People's Republic of China
| | - Shijun Hong
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, People's Republic of China
| | - Yi Ye
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xueyan Wang
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fan Chen
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lin Yang
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Youyi Yan
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Linchuan Liao
- Department of Analytical Toxicology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
17
|
Zhao Z, Li B, Wu Y, Chen X, Guo Y, Shen Y, Huang H. Ketamine affects the integration of developmentally generated granule neurons in the adult stage. BMC Neurosci 2019; 20:60. [PMID: 31852437 PMCID: PMC6921590 DOI: 10.1186/s12868-019-0542-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/07/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Ketamine has been reported to cause neonatal neurotoxicity in a variety of developing animal models. Various studies have been conducted to study the mechanism of neurotoxicity for general anesthetic use during the neonatal period. Previous experiments have suggested that developmentally generated granule neurons in the hippocampus dentate gyrus (DG) supported hippocampus-dependent memory. Therefore, this study aimed to investigate whether ketamine affects the functional integration of developmentally generated granule neurons in the DG. For this purpose,the postnatal day 7 (PND-7) Sprague-Dawley (SD) rats were divided into the control group and the ketamine group (rats who received 4 injections of 40 mg/kg ketamine at 1 h intervals). To label dividing cells, BrdU was administered for three consecutive days after the ketamine exposure; NeuN+/BrdU+cells were observed by using immunofluorescence. To evaluate the developmentally generated granule neurons that support hippocampus-dependent memory, spatial reference memory was tested by using Morris Water Maze at 3 months old, after which the immunofluorescence was used to detect c-Fos expression in the NeuN+/BrdU+ cells. The expression of caspase-3 was measured by western blot to detect the apoptosis in the hippocampal DG. RESULTS The present results showed that the neonatal ketamine exposure did not influence the survival rate of developmentally generated granule neurons at 2 and 3 months old, but ketamine interfered with the integration of these neurons into the hippocampal DG neural circuits and caused a deficit in hippocampal-dependent spatial reference memory tasks. CONCLUSIONS In summary, these findings may promote more studies to investigate the neurotoxicity of ketamine in the developing brain.
Collapse
Affiliation(s)
- Zhanqiang Zhao
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Bing Li
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China
| | - Xujun Chen
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yan Guo
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yang Shen
- Department of Anesthesiology, Jiangning Hospital of Traditional Chinese Medicine, Nanjing, China
| | - He Huang
- Department of Anesthesiology, First Affiliated Hospital With Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
18
|
Chen YC, Sheen JM, Hsu MH, Hsiao CC, Wang SC, Huang LT. Melatonin rescued methotrexate-induced spatial deficit and hyperhomocysteinemia and increased asymmetric dimethylarginine in plasma and dorsal hippocampus in developing rats. Life Sci 2019; 242:116931. [PMID: 31618610 DOI: 10.1016/j.lfs.2019.116931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 01/08/2023]
Abstract
AIMS With the improvement of the survival rates in children acute lymphoblastic leukemia (ALL), some children ALL survivors show impaired cognitive function. Methotrexate (MTX), an essential component in ALL treatment, has been reported to be related to neurologic sequelae and to increased oxidative stress through its interactions with enzymes in the folate pathway. Asymmetric dimethylarginine (ADMA) is the main endogenous inhibitor of nitric oxide synthase, and increased ADMA may result from increased oxidants. Melatonin is an antioxidant; however, its role in MTX neuropathy is not well studied. We developed a rat model mimicking child ALL treatment to explore peripheral and central homocysteine and ADMA regulation after MTX and found potential treatment choice. MAIN METHODS Preweaning male Sprague-Dawley rats were used in this study. Experiment 1 evaluated spatial performance in rats with intrathecal (IT) MTX, intraperitoneal (IP) MTX, or combined IT and IP MTX, protocols mimicking ALL treatment in children. Experiment 2 focused on rats with combined IT and IP MTX, evaluating spatial performance and plasma and dorsal hippocampal homocysteine and ADMA levels, their regulation, and the protective effect of melatonin. KEY FINDINGS Combined IT and IP MTX treatment caused in spatial deficits in developing rats, and melatonin restored the spatial performance. Alterations in peripheral and central homocysteine and ADMA concentrations and their regulation were found and could be alleviated by melatonin treatment. SIGNIFICANCES Combined IP and IT MTX treatment caused spatial deficits in developing rats. Melatonin could restore spatial performance through alleviating the effects on the imbalance of oxidative stress.
Collapse
Affiliation(s)
- Yu-Chieh Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Traditional Medicine, Chang Gung University, Linkow, Taiwan
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Traditional Medicine, Chang Gung University, Linkow, Taiwan
| | - Mei-Hsin Hsu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Cheng Hsiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Traditional Medicine, Chang Gung University, Linkow, Taiwan
| | - Su-Chen Wang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Traditional Medicine, Chang Gung University, Linkow, Taiwan.
| |
Collapse
|
19
|
Li W, Li H, Wei H, Lu Y, Lei S, Zheng J, Lu H, Chen X, Liu Y, Zhang P. 17β-Estradiol Treatment Attenuates Neurogenesis Damage and Improves Behavior Performance After Ketamine Exposure in Neonatal Rats. Front Cell Neurosci 2019; 13:251. [PMID: 31263401 PMCID: PMC6585163 DOI: 10.3389/fncel.2019.00251] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 05/20/2019] [Indexed: 12/23/2022] Open
Abstract
Ketamine exposure disturbed normal neurogenesis in the developing brain and resulted in subsequent neurocognitive deficits. 17β-estradiol provides robust neuroprotection in a variety of brain injury models in animals of both sexes and attenuates neurodegeneration induced by anesthesia agents. In the present study, we aimed to investigate whether 17β-estradiol could attenuate neonatal ketamine exposure-disturbed neurogenesis and behavioral performance. We treated 7-day-old (Postnatal day 7, PND 7) Sprague-Dawley rats and neural stem cells (NSCs) with either normal saline, ketamine, or 17β-estradiol before/after ketamine exposure, respectively. At PND 14, the rats were decapitated to detect neurogenesis in the subventricular zone (SVZ) and subgranular zone (SGZ) of the hippocampus by immunofluorescence staining. The proliferation, neuronal differentiation, and apoptosis of NSCs were assessed by immunohistochemistry method and TUNEL assay, respectively. The protein levels of cleaved caspase-3 in vivo in addition to GSK-3β and p-GSK-3β in vitro were examined by western blotting. Spatial learning and memory abilities were assessed by Morris water maze (MWM) test at PND 42–47. Ketamine exposure decreased cell proliferation in the SVZ and SGZ, inhibited NSC proliferation and neuronal differentiation, promoted NSC apoptosis and led to adult cognitive deficits. Furthermore, ketamine increased cleaved caspase-3 in vivo and decreased the expression of p-GSK-3β in vitro. Treatment with 17β-estradiol could attenuate ketamine-induced changes both in vivo and in vitro. For the first time we showed that 17β-estradiol alleviated ketamine-induced neurogenesis inhibition and cognitive dysfunction in the developing rat brain. Moreover, the protection of 17β-estradiol was associated with GSK-3β.
Collapse
Affiliation(s)
- Weisong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huixian Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Lei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Juan Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haixia Lu
- Institute of Neurobiology, National Key Academic Subject, Physiology of Xi'an Jiaotong University, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, National Key Academic Subject, Physiology of Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, National Key Academic Subject, Physiology of Xi'an Jiaotong University, Xi'an, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
20
|
Michaëlsson H, Andersson M, Svensson J, Karlsson L, Ehn J, Culley G, Engström A, Bergström N, Savvidi P, Kuhn H, Hanse E, Seth H. The novel antidepressant ketamine enhances dentate gyrus proliferation with no effects on synaptic plasticity or hippocampal function in depressive-like rats. Acta Physiol (Oxf) 2019; 225:e13211. [PMID: 30347138 DOI: 10.1111/apha.13211] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/18/2022]
Abstract
AIM Major depressive disorder is a common and debilitating condition with substantial economic impact. Treatment options, although effective, are aimed at relieving the symptoms with limited disease modification. Ketamine, a commonly used anaesthetic, has received substantial attention as it shows rapid antidepressant effects clinically. We studied the effects of ketamine on hippocampal function and dentate gyrus proliferation in rats showing a depressive-like phenotype. METHODS Adolescent and adult animals were pre-natally exposed to the glucocorticoid analog dexamethasone, and we verified a depressive-like phenotype using behavioural tests, such as the sucrose preference. We subsequently studied the effects of ketamine on hippocampal synaptic transmission, plasticity and dentate gyrus proliferation. In addition, we measured hippocampal glutamate receptor expression. We also tested the ketamine metabolite hydroxynorketamine for NMDA-receptor independent effects. RESULTS Surprisingly, our extensive experimental survey revealed limited effects of ketamine or its metabolite on hippocampal function in control as well as depressive-like animals. We found no effects on synaptic efficacy or induction of long-term potentiation in adolescent and adult animals. Also there was no difference when comparing the dorsal and ventral hippocampus. Importantly, however, ketamine 24 hours prior to experimentation significantly increased the dentate gyrus proliferation, as revealed by Ki-67 immunostaining, in the depressive-like phenotype. CONCLUSION We find limited effects of ketamine on hippocampal glutamatergic transmission. Instead, alterations in dentate gyrus proliferation could explain the antidepressant effects of ketamine.
Collapse
Affiliation(s)
- Henrik Michaëlsson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Mats Andersson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Johan Svensson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Lars Karlsson
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Johan Ehn
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Georgia Culley
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Anders Engström
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Nicklas Bergström
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Parthenia Savvidi
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Hans‐Georg Kuhn
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Eric Hanse
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| | - Henrik Seth
- Department of Neuroscience and Physiology University of Gothenburg Gothenburg Sweden
| |
Collapse
|
21
|
Scott L, Downing T. Dose-Dependent Adult Neurodegeneration in a Rat Model After Neonatal Exposure to β-N-Methylamino-l-Alanine. Neurotox Res 2019; 35:711-723. [DOI: 10.1007/s12640-019-9996-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/27/2018] [Accepted: 01/08/2019] [Indexed: 01/18/2023]
|
22
|
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
|
23
|
Baghishani F, Mohammadipour A, Hosseinzadeh H, Hosseini M, Ebrahimzadeh-Bideskan A. The effects of tramadol administration on hippocampal cell apoptosis, learning and memory in adult rats and neuroprotective effects of crocin. Metab Brain Dis 2018; 33:907-916. [PMID: 29470767 PMCID: PMC5956046 DOI: 10.1007/s11011-018-0194-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 01/22/2018] [Indexed: 02/07/2023]
Abstract
Tramadol, a frequently used pain reliever drug, present neurotoxic effects associated to cognitive dysfunction. Moreover, crocin has been reported to have neuroprotective effects. The aim of this study was to assess crocin's capacity to protect learning, and memory abilities on tramadol-treated rats. A total of 35 rats were divided into five groups: Control, Saline, tramadol (50 mg/kg), tramadol + crocin(30 mg/kg), crocin groups and treated orally for 28 consecutive days. Morris water maze (MWM) and passive avoidance (PA) tests were done, followed by dissection of the rat's brains for toluidine blue and TUNEL staining. In MWM test, tramadol group spent lower time and traveled shorter distance in the target quadrant (Q1) (P < 0.05). On the other side, the traveled distance in tramadol-crocin group was higher than tramadol (P < 0.05). In PA test, both the delay for entering the dark, and the total time spent in the light compartment decreased in tramadol comparing to the control group (P < 0.05), while it increased in tramadol-crocin compared with the tramadol group (P < 0.05). In tramadol-treated animals, the dark neurons (DNs) and apoptotic cells in CA1, CA3 and DG increased (P < 0.05), while concurrent intake of crocin decreased the number of DNs and apoptotic cells in these areas (P < 0.05). Crocin was able to improve learning and memory of tramadol-treated rats and also decreased DNs and apoptotic cells in the hippocampus. Considering these results, the potential capacity of crocin for decreasing side effects of tramadol on the nervous system is suggested.
Collapse
Affiliation(s)
- Farideh Baghishani
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran
| | - Abbas Mohammadipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran
- Microanatomy Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossain Hosseinzadeh
- Pharmaceutical Research Center, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran.
- Microanatomy Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
24
|
Chamaa F, Bahmad HF, Makkawi AK, Chalhoub RM, Al-Chaer ED, Bikhazi GB, Nahas Z, Abou-Kheir W. Nitrous Oxide Induces Prominent Cell Proliferation in Adult Rat Hippocampal Dentate Gyrus. Front Cell Neurosci 2018; 12:135. [PMID: 29867368 PMCID: PMC5967150 DOI: 10.3389/fncel.2018.00135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 04/30/2018] [Indexed: 12/28/2022] Open
Abstract
The identification of distinct and more efficacious antidepressant treatments is highly needed. Nitrous oxide (N2O) is an N-methyl-D-aspartic acid (NMDA) antagonist that has been reported to exhibit antidepressant effects in treatment-resistant depression (TRD) patients. Yet, no studies have investigated the effects of sub-anesthetic dosages of N2O on hippocampal cell proliferation and neurogenesis in adult brain rats. In our study, adult male Sprague-Dawley rats were exposed to single or multiple exposures to mixtures of 70% N2O and 30% oxygen (O2). Sham groups were exposed to 30% O2 and the control groups to atmospheric air. Hippocampal cell proliferation was assessed by bromodeoxyuridine (BrdU) incorporation, and BrdU-positive cells were counted in the dentate gyrus (DG) using confocal microscopy. Results showed that while the rates of hippocampal cell proliferation were comparable between the N2O and sham groups at day 1, levels increased by 1.4 folds at day 7 after one session exposure to N2O. Multiple N2O exposures significantly increased the rate of hippocampal cell proliferation to two folds. Therefore, sub-anesthetic doses of N2O, similar to ketamine, increase hippocampal cell proliferation, suggesting that there will ultimately be an increase in neurogenesis. Future studies should investigate added N2O exposures and their antidepressant behavioral correlates.
Collapse
Affiliation(s)
- Farah Chamaa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hisham F. Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ahmad-Kareem Makkawi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Reda M. Chalhoub
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Elie D. Al-Chaer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George B. Bikhazi
- Department of Anesthesiology, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ziad Nahas
- Department of Psychiatry, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Department of Psychiatry, Medical School, University of Minnesota, Minneapolis, MN, United States
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
25
|
Zuo D, Liu Y, Liu Z, Cui J, Zhou X, Liu Y, Li Z, Wu Y. Alcohol aggravates ketamine-induced behavioral, morphological and neurochemical alterations in adolescent rats: The involvement of CREB-related pathways. Behav Brain Res 2018; 349:80-90. [PMID: 29738804 DOI: 10.1016/j.bbr.2018.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 01/03/2023]
Abstract
Currently, an increasing proportion of adolescent ketamine users simultaneously consume alcohol. However, the potential behavioural and neurological alterations induced by such a drug combination and the underlying mechanisms have not been systematically examined. Therefore, in the present study, the behavioural and morphological changes and the underlying mechanisms were studied in adolescent rats after repeated alcohol and/or ketamine treatment. This study provided the first evidence that co-administration of alcohol (2 and 4 g/kg, i.g.) in adolescent rats significantly potentiated the neurotoxic properties of repeated ketamine (30 mg/kg, i.p.) treatments over 14 days, manifesting as increased locomotor activity, stereotypic behaviour, ataxia and morphological changes. This potentiation was associated with the enhancement by alcohol of ketamine-induced glutamate (Glu) and dopamine (DA) release in the cortex and hippocampus. Further mechanistic study demonstrated that alcohol potentiated ketamine-induced neurotoxicity through down-regulation of Akt (a serine/threonine kinase or protein kinase, PKB), protein kinase A (PKA), calmodulin-dependent kinase IV (CaMK-IV)-mediated cyclic AMP-responsive element binding protein (CREB) pathways and induction of neuronal apoptosis in the cortex and hippocampus of the adolescent rats. As this study provides strong evidence that repeated alcohol and ketamine co-exposure may cause serious neurotoxicity, attention needs to be drawn to the potential risk of this consumption behaviour, especially for adolescents.
Collapse
Affiliation(s)
- Daiying Zuo
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Yumiao Liu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zi Liu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Jiahui Cui
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Xuejiao Zhou
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Yang Liu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zengqiang Li
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Yingliang Wu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
26
|
Protective effects of pituitary adenylate cyclase activating polypeptide against neurotoxic agents. Neurotoxicology 2018; 66:185-194. [DOI: 10.1016/j.neuro.2018.03.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 03/09/2018] [Accepted: 03/27/2018] [Indexed: 01/28/2023]
|
27
|
Abstract
PURPOSE OF REVIEW This article reviews the most recently published evidence that investigated anesthesia-induced neurotoxicity in both animals and humans, especially as it pertains to the perinatal period. RECENT FINDINGS Several recent studies have focused on better understanding the complex mechanisms that underlie intravenous and volatile anesthesia-induced neurotoxicity in animals. Adjuvant agents that target these pathways have been investigated for their effectiveness in attenuating the neuroapoptosis and neurocognitive deficits that result from anesthesia exposure, including dexmedetomidine, rutin, vitamin C, tumor necrosis factor α, lithium, apocynin, carreic acid phenethyl ester. Five clinical studies, including one randomized control trial, provided inconsistent evidence on anesthesia-induced neurotoxicity in humans. SUMMARY Despite a growing body of preclinical studies that have demonstrated anesthesia-induced neurotoxic effects in the developing and aging brain, their effects on the human brain remains to be determined. The performance of large-scale human studies is limited by several important factors, and noninvasive biomarkers and neuroimaging modalities should be employed to define the injury phenotypes that reflect anesthesia-induced neurotoxicity. Ultimately, the use of these modalities may provide new insights into whether the concerns of anesthetics are justified in humans.
Collapse
|
28
|
Zhou X, Zhang P, Luo W, Zhang L, Hu R, Sun Y, Jiang H. Ketamine induces apoptosis in lung adenocarcinoma cells by regulating the expression of CD69. Cancer Med 2018; 7:788-795. [PMID: 29453833 PMCID: PMC5852367 DOI: 10.1002/cam4.1288] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/17/2017] [Accepted: 11/01/2017] [Indexed: 01/14/2023] Open
Abstract
Ketamine, an anesthetic, analgesic, or sedative, is widely used for the treatment of cancer pain. Recently, ketamine has been also reported to be tumor repressor for inhibiting proliferation, invasion, and migration, and inducing apoptosis in many cancers. However, whether ketamine can induce the apoptosis of lung adenocarcinoma (LUAD) and which downstream molecular mediates its function remain largely unknown. A LUAD cell line A549 was incubated with ketamine at 0, 1, 10, and 100 μmol/L for 24 h. Trypan blue staining was used to detect the cell viability. Flow cytometry (FACS) was applied to evaluate cell apoptosis proportion. The expression of CD69 was quantitated by western blotting. Ketamine induced the A549 cell apoptosis in a concentration-dependent manner. CD69 was downregulated in LUAD patients' cancer tissue compared with the normal tissue. CD69 can be upregulated in ketamine treating A549 cells and induce the A549 cell apoptosis. Rescue experiment showed that downregulation of CD69 significantly blocked the function of ketamine on inducing apoptosis. Taken together, our results demonstrated that ketamine induced LUAD cells apoptosis by upregulating the CD69 expression. This study suggests that the ketamine can be potential drug for LUAD treatment, and the ketamine/CD69 signaling may be the new potential therapeutic target LUAD therapy.
Collapse
Affiliation(s)
- Xuhui Zhou
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, 200011, China
| | - Peihong Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, 200011, China
| | - Wei Luo
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, 200011, China
| | - Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, 200011, China
| | - Rong Hu
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, 200011, China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, 200011, China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, 200011, China
| |
Collapse
|
29
|
Ivan Ezquerra-Romano I, Lawn W, Krupitsky E, Morgan CJA. Ketamine for the treatment of addiction: Evidence and potential mechanisms. Neuropharmacology 2018; 142:72-82. [PMID: 29339294 DOI: 10.1016/j.neuropharm.2018.01.017] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/08/2018] [Accepted: 01/11/2018] [Indexed: 12/14/2022]
Abstract
Ketamine is a dissociative anaesthetic drug which acts on the central nervous system chiefly through antagonism of the n-methyl-d-aspartate (NMDA) receptor. Recently, ketamine has attracted attention as a rapid-acting anti-depressant but other studies have also reported its efficacy in reducing problematic alcohol and drug use. This review explores the preclinical and clinical research into ketamine's ability to treat addiction. Despite methodological limitations and the relative infancy of the field, results thus far are promising. Ketamine has been shown to effectively prolong abstinence from alcohol and heroin in detoxified alcoholics and heroin dependent individuals, respectively. Moreover, ketamine reduced craving for and self-administration of cocaine in non-treatment seeking cocaine users. However, further randomised controlled trials are urgently needed to confirm ketamine's efficacy. Possible mechanisms by which ketamine may work within addiction include: enhancement of neuroplasticity and neurogenesis, disruption of relevant functional neural networks, treating depressive symptoms, blocking reconsolidation of drug-related memories, provoking mystical experiences and enhancing psychological therapy efficacy. Identifying the mechanisms by which ketamine exerts its therapeutic effects in addiction, from the many possible candidates, is crucial for advancing this treatment and may have broader implications understanding other psychedelic therapies. In conclusion, ketamine shows great promise as a treatment for various addictions, but well-controlled research is urgently needed. This article is part of the Special Issue entitled 'Psychedelics: New Doors, Altered Perceptions'.
Collapse
Affiliation(s)
- I Ivan Ezquerra-Romano
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - W Lawn
- Clinical Psychopharmacology Unit, University College London, Gower Street, London, UK
| | - E Krupitsky
- St.-Petersburg Pavlov State Medical University and Bekhterev Research Psychoneurological Institute, St. Petersburg, Russia
| | - C J A Morgan
- Clinical Psychopharmacology Unit, University College London, Gower Street, London, UK; Psychopharmacology and Addiction Research Centre, University of Exeter, Exeter, UK.
| |
Collapse
|
30
|
Advantages of the Alpha-lipoic Acid Association with Chlorpromazine in a Model of Schizophrenia Induced by Ketamine in Rats: Behavioral and Oxidative Stress evidences. Neuroscience 2018; 373:72-81. [PMID: 29337238 DOI: 10.1016/j.neuroscience.2018.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/27/2017] [Accepted: 01/03/2018] [Indexed: 11/20/2022]
Abstract
Schizophrenia is a chronic mental disorder reported to compromise about 1% of the world's population. Although its pathophysiological process is not completely elucidated, evidence showing the presence of an oxidative imbalance has been increasingly highlighted in the literature. Thus, the use of antioxidant substances may be of importance for schizophrenia treatment. The objective of this study was to evaluate the behavioral and oxidative alterations by the combination of chlorpromazine (CP) and alpha-lipoic acid (ALA), a potent antioxidant, in the ketamine (KET) model of schizophrenia in rats. Male Wistar rats (200-300 g) were treated for 10 days with saline, CP or ALA alone or in combination with CP previous to KET and the behavioral (open field, Y-maze and PPI tests) and oxidative tests were performed on the last day of treatment. The results showed that KET induced hyperlocomotion, impaired working memory and decreased PPI. CP alone or in combination with ALA prevented KET-induced behavioral effects. In addition, the administration of KET decreased GSH and increased nitrite, lipid peroxidation and myeloperoxidase activity. CP alone or combined with ALA prevented the oxidative alterations induced by KET. In conclusion, the treatment with KET in rats induced behavioral impairments accompanied by hippocampal oxidative alterations, possibly related to NMDA receptors hypofunction. Besides that, CP alone or combined with ALA prevented these effects, showing a beneficial activity as antipsychotic agents.
Collapse
|
31
|
Scott LL, Downing TG. A Single Neonatal Exposure to BMAA in a Rat Model Produces Neuropathology Consistent with Neurodegenerative Diseases. Toxins (Basel) 2017; 10:E22. [PMID: 29286334 PMCID: PMC5793109 DOI: 10.3390/toxins10010022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/23/2017] [Accepted: 12/27/2017] [Indexed: 12/11/2022] Open
Abstract
Although cyanobacterial β-N-methylamino-l-alanine (BMAA) has been implicated in the development of Alzheimer's Disease (AD), Parkinson's Disease (PD) and Amyotrophic Lateral Sclerosis (ALS), no BMAA animal model has reproduced all the neuropathology typically associated with these neurodegenerative diseases. We present here a neonatal BMAA model that causes β-amyloid deposition, neurofibrillary tangles of hyper-phosphorylated tau, TDP-43 inclusions, Lewy bodies, microbleeds and microgliosis as well as severe neuronal loss in the hippocampus, striatum, substantia nigra pars compacta, and ventral horn of the spinal cord in rats following a single BMAA exposure. We also report here that BMAA exposure on particularly PND3, but also PND4 and 5, the critical period of neurogenesis in the rodent brain, is substantially more toxic than exposure to BMAA on G14, PND6, 7 and 10 which suggests that BMAA could potentially interfere with neonatal neurogenesis in rats. The observed selective toxicity of BMAA during neurogenesis and, in particular, the observed pattern of neuronal loss observed in BMAA-exposed rats suggest that BMAA elicits its effect by altering dopamine and/or serotonin signaling in rats.
Collapse
Affiliation(s)
- Laura Louise Scott
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| | - Timothy Grant Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| |
Collapse
|
32
|
Scott LL, Downing TG. Β-N-Methylamino-L-Alanine (BMAA) Toxicity Is Gender and Exposure-Age Dependent in Rats. Toxins (Basel) 2017; 10:E16. [PMID: 29280981 PMCID: PMC5793103 DOI: 10.3390/toxins10010016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/21/2017] [Accepted: 12/26/2017] [Indexed: 12/21/2022] Open
Abstract
Cyanobacterial β-N-methylamino-L-alanine (BMAA) has been suggested as a causative or contributory factor in the development of several neurodegenerative diseases. However, no BMAA animal model has adequately shown clinical or behavioral symptoms that correspond to those seen in either Alzheimer's Disease (AD), Amyotrophic Lateral Sclerosis (ALS) or Parkinson's Disease (PD). We present here the first data that show that when neonatal rats were exposed to BMAA on postnatal days 3, 4 and 5, but not on gestational day 14 or postnatally on days 7 or 10, several AD and/or PD-related behavioral, locomotor and cognitive deficits developed. Male rats exhibited severe unilateral hindlimb splay while whole body tremors could be observed in exposed female rats. BMAA-exposed rats failed to identify and discriminate a learned odor, an early non-motor symptom of PD, and exhibited decreased locomotor activity, decreased exploration and increased anxiety in the open field test. Alterations were also observed in the rats' natural passive defense mechanism, and potential memory deficits and changes to the rat's natural height avoidance behavior could be observed as early as PND 30. Spatial learning, short-term working, reference and long-term memory were also impaired in 90-day-old rats that had been exposed to a single dose of BMAA on PND 3-7. These data suggest that BMAA is a developmental neurotoxin, with specific target areas in the brain and spinal cord.
Collapse
Affiliation(s)
- Laura Louise Scott
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| | - Timothy Grant Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| |
Collapse
|
33
|
Ye Z, Li Q, Guo Q, Xiong Y, Guo D, Yang H, Shu Y. Ketamine induces hippocampal apoptosis through a mechanism associated with the caspase-1 dependent pyroptosis. Neuropharmacology 2017; 128:63-75. [PMID: 28963039 DOI: 10.1016/j.neuropharm.2017.09.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/24/2017] [Accepted: 09/25/2017] [Indexed: 12/11/2022]
Abstract
Ketamine, a pediatric anesthetic, is widely used in clinical practice. There was growing evidence showing that ketamine can promote neuronal death in developing brains of both humans and animals. In this study, we used in vivo neonatal and juvenile mouse models to induce ketamine-related neurotoxicity in the hippocampus. Active caspase-3 and -9 proteins, which are responsible for the release of cytochrome C, and the mitochondrial translocation of p53, which is associated with mitochondrial apoptosis, were found to be significantly up-regulated in the ketamine-induced hippocampal neurotoxicity. Furthermore, we demonstrated that the levels of pyroptosis-related proteins, including caspase-1 and -11, NOD-like receptor family, pyrin domain containing 3 (NLRP3), and IL-1β and IL-18, significantly increased after multiple doses of ketamine administration. We speculated that ketamine triggered the formation of NLRP3 and caspase-1 complex and its translocation to the mitochondria. In consistent with this, ketamine treatment was found to induce pyroptosis in mouse primary hippocampal neurons, which was characterized by increased pore formation and elevated lactate dehydrogenase release in mitochondria. Silencing caspase-1 with lentivirus-mediated short hairpin RNA (shRNA) significantly decreased the levels of not only pyroptosis-related proteins but also mitochondrial apoptosis-associated proteins in mouse primary hippocampal neurons. We conclude that caspase-1-dependent pyroptosis is an important event which may be an essential pathway involved in the mitochondria-associated apoptosis in ketamine-induced hippocampal neurotoxicity.
Collapse
Affiliation(s)
- Zhi Ye
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA; Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Qing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA; Institute of Clinical Pharmacology, Central South University, Hunan 410078, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Yunchuan Xiong
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Dong Guo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA
| | - Hong Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA.
| |
Collapse
|
34
|
Huang H, Liu CM, Sun J, Jin WJ, Wu YQ, Chen J. Repeated 2% sevoflurane administration in 7‑ and 60-day-old rats : Neurotoxicity and neurocognitive dysfunction. Anaesthesist 2017; 66:850-857. [PMID: 28914327 DOI: 10.1007/s00101-017-0359-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Sevoflurane is one of the most widely used inhalation anesthetics in pediatric anesthesia. A large number of studies have demonstrated that repeated treatment with high concentrations or long durations of sevoflurane anesthesia during the neonatal period can induce neuroapoptosis and long-term learning disability. In clinical practice, we observed that a subset of patients underwent minor surgery under sevoflurane anesthesia more than once from birth to adolescence. Therefore, this research was conducted to investigate whether a 2% concentration of sevoflurane (clinically relevant usage of sevoflurane) for 1 h (a short duration) can induce neuroapoptosis and neurocognitive dysfunction in adolescent rats that received sevoflurane (2% for 1 h) during the neonatal period. MATERIAL AND METHODS Group I: neonatal rats at postnatal day 7 (PND-7) were treated with oxygen under controlled conditions and then raised to PND-60. Group II: PND-7 rats were treated with 2% sevoflurane for 1 h and then raised to PND-60. Group III: the PND-60 rats were treated with 2% sevoflurane for 1 h and in group IV the PND-7 rats were treated with 2% sevoflurane for 1 h and then anesthetized with 2% sevoflurane for 1 h at PND-60 again. The expression of caspase-3, Bax and Bcl-2 in the hippocampal dentate gyrus (DG) were measured by Western blot analysis. Neuroapoptosis in the hippocampal DG was assessed using NeuN/caspase-3 double-immunofluorescence staining. Spatial reference memory was tested by the Morris water maze test. RESULTS The present data showed that sevoflurane (2% for 1 h) did not induce obvious hippocampal neuroapoptosis in the PND-7 rats and PND-60 rats; their performance in hippocampal-dependent spatial memory was not significantly impaired; however, the rats in group IV showed poor performance in the Morris water maze test and the neuroapoptosis in group IV was significantly increased. CONCLUSION Our findings suggested that sevoflurane can induce neuroapoptosis and cognitive dysfunction in adolescent rats that received repeated sevoflurane (2% for 1 h) during the postnatal period. These findings will promote further studies to investigate the effects of repeated sevoflurane exposure on the development of the central nervous system and function of learning and memory, as well as the underlying mechanisms in vitro and in vivo.
Collapse
Affiliation(s)
- He Huang
- Department of Anesthesiology, The First Affiliated Hospital with Nanjing Medical University, Guangzhou Road 300, 210029, Nanjing, China
| | - Cun-Ming Liu
- Department of Anesthesiology, The First Affiliated Hospital with Nanjing Medical University, Guangzhou Road 300, 210029, Nanjing, China
| | - Jie Sun
- Department of Anesthesiology, The First Affiliated Hospital with Nanjing Medical University, Guangzhou Road 300, 210029, Nanjing, China
| | - Wen-Jie Jin
- Department of Anesthesiology, The First Affiliated Hospital with Nanjing Medical University, Guangzhou Road 300, 210029, Nanjing, China
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, China
| | - Jing Chen
- Department of Anesthesiology, The First Affiliated Hospital with Nanjing Medical University, Guangzhou Road 300, 210029, Nanjing, China.
| |
Collapse
|
35
|
Alcohol amplifies ketamine-induced apoptosis in primary cultured cortical neurons and PC12 cells through down-regulating CREB-related signaling pathways. Sci Rep 2017; 7:10523. [PMID: 28874724 PMCID: PMC5585325 DOI: 10.1038/s41598-017-10868-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 08/16/2017] [Indexed: 12/22/2022] Open
Abstract
Recreational use of ketamine (KET) has been increasing worldwide. Previous studies have demonstrated that KET induced neurotoxicity; however, few studies have examined how alcohol (ALC) affects KET-induced neurotoxicity. In light of the fact that some KET abusers combine KET with ALC, the present study was aimed to investigate the effects of ALC on KET-induced neurotoxicity and the underlying mechanism in vitro. Our data revealed that co-treatment with ALC and KET was more detrimental to cell viability than KET single treatment in both PC12 cells and primary cultured rat cortical neurons. Furthermore, ALC exacerbated KET-induced apoptosis characterized by morphological changes and the sub-G1 phase increase, which were mitigated by the pretreatment of CNQX, a known alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA)/kainite (KA) receptor antagonist. In addition, ALC and KET co-treatment led to intracellular Ca2+ overload, down-regulation of p-Akt, p-CREB, PKA, CaMK-IV, Bcl-2 and BDNF expression and up-regulation of cleaved caspase-3 and Bax expression, which can be attenuated by CNQX pretreatment. These results indicate that the potentiation of ALC on KET-induced neurotoxicity was related to the down-regulation of CREB-related pathways. Our present study also indicates that ALC and KET co-abuse might cause serious neurotoxicity which should be conveyed to the public and drew enough attention.
Collapse
|
36
|
Li Y, Shen R, Wen G, Ding R, Du A, Zhou J, Dong Z, Ren X, Yao H, Zhao R, Zhang G, Lu Y, Wu X. Effects of Ketamine on Levels of Inflammatory Cytokines IL-6, IL-1β, and TNF-α in the Hippocampus of Mice Following Acute or Chronic Administration. Front Pharmacol 2017; 8:139. [PMID: 28373844 PMCID: PMC5357631 DOI: 10.3389/fphar.2017.00139] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/06/2017] [Indexed: 12/20/2022] Open
Abstract
Ketamine is an injectable anesthetic and recreational drug of abuse commonly used worldwide. Many experimental studies have shown that ketamine can impair cognitive function and induce psychotic states. Neuroinflammation has been suggested to play an important role in neurodegeneration. Meanwhile, ketamine has been shown to modulate the levels of inflammatory cytokines. We hypothesized that the effects of ketamine on the central nervous system are associated with inflammatory cytokines. Therefore, we set out to establish acute and chronic ketamine administration models in C57BL/6 mice, to evaluate spatial recognition memory and emotional response, to analyze the changes in the levels of the inflammatory cytokines interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) in the mouse hippocampus, employing behavioral tests, Western blot, quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and immunohistochemistry. Our results showed that ketamine at the dose of 60 mg/kg induced spatial recognition memory deficit and reduced anxiety-like behaviors in mice after chronic administration. Moreover, we found that ketamine increased the hippocampal levels of IL-6 and IL-1β after single, multiple and long-term administration in a dose-dependent manner. However, the expression level of TNF-α differed in the mouse hippocampus under different conditions. Single administration of ketamine increased the level of TNF-α, whereas multiple and long-term administration decreased it significantly. We considered that TNF-α expression could be controlled by a bi-directional regulatory pathway, which was associated with the dose and duration of ketamine administration. Our results suggest that the alterations in the levels of inflammatory cytokines IL-6, IL-1β, and TNF-α may be involved in the neurotoxicity of ketamine.
Collapse
Affiliation(s)
- Yanning Li
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Ruipeng Shen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
- Wujiang District Branch of Suzhou Public Security Bureau, SuzhouChina
| | - Gehua Wen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Runtao Ding
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Ao Du
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Jichuan Zhou
- Key Laboratory of Health Ministry in Congenital Malformation, The Affiliated Shengjing Hospital of China Medical University, ShenyangChina
| | - Zhibin Dong
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Xinghua Ren
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Hui Yao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Rui Zhao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Guohua Zhang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, The Affiliated Shengjing Hospital of China Medical University, ShenyangChina
| | - Xu Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, ShenyangChina
| |
Collapse
|
37
|
Influence of General Anesthesia on Impulsivity and Learning Ability-Experimental Study. ACTA MEDICA MARISIENSIS 2016. [DOI: 10.1515/amma-2016-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objective: To investigate the effect of anesthesia on rats’ ability of learning and over their impulsivity.
Material and Methods: We studied eight Wistar adult male rats, test and drug naive subjects. Animals were separated in two groups, group A and B with four members each. Group A included the anesthetized animals. The combination of ketamine, xylazine and piplophen in 2ml/kg body weight dosage was used and testing was done 24 hours after anesthesia. Group B was taken as control. The study was conducted using the ”Delay discounting” apparatus. Experiments assessing impulsive behavior were conducted using automated operant chambers, equipped with two nose-poke holes (holes where pellets of food were released). Rat’s answer was considered touching the nose-poke hole. One answer was rewarded with pellets of food of 45 mg each (small reward), while another hole released five pellets of 45 mg each (high reward). Both types of rewards were presented immediately after rat’s answer and were followed for a period of 25 seconds timeout. During the training phase, rats were placed in operant chambers 30 minutes per day, 5 consecutive days. The growing percent of preference for greater reward indicates learning. For the testing phase the procedure was similar, but a delay was introduced before the release of the big reward. During this phase, the preference for higher reward was indicative for non-impulsive behaviour.
Results: The results didn’t show significant statistically differences between the two groups.
Conclusions: Anesthesia had no effect on learning ability nor on impulsivity.
Collapse
|
38
|
Chronic stress prior to pregnancy potentiated long-lasting postpartum depressive-like behavior, regulated by Akt-mTOR signaling in the hippocampus. Sci Rep 2016; 6:35042. [PMID: 27756905 PMCID: PMC5069466 DOI: 10.1038/srep35042] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 09/19/2016] [Indexed: 12/14/2022] Open
Abstract
Postpartum depression (PPD) affects over 10% of new mothers and adversely impacts the health of offspring. One of the greatest risk factors for PPD is prepregnancy stress but the underlying biological mechanism is unknown. Here we constructed an animal model which recapitulated prepregnancy stress induced PPD and tested the role of Akt-mTOR signaling in the hippocampus. Female virgin Balb/c mice received chronic restraint stress, followed by co-housing with a normal male mouse. We found that the chronic stress led to a transient depressive-like condition that disappeared within two weeks. However, prepregnantly stressed females developed long-term postpartum depressive-like (PPD-like) symptoms as indicated by deficient performance in tests of sucrose preference, forced swim, and novelty-suppressed feeding. Chronic stress induced transient decrease in Akt-mTOR signaling and altered expressions of glutamate receptor subunits NR1 and GluR1, in contrast to long-term deficits in Akt-mTOR signaling, GluR1/NR1 ratio, and hippocampal neurogenesis in PPD-like mice. Acute ketamine improved the molecular signaling abnormality, and reversed the behavioral deficits in PPD-like mice in a rapid and persistent manner, in contrast to ineffectiveness by chronic fluoxetine treatment. Taken together, we find that chronic prepregnancy stress potentiates a long-term PPD, in which Akt-mTOR signaling may play a crucial role.
Collapse
|