1
|
Huang XM, Qiu HX. Effect of intravenous esketamine in emergency cesarean deliveries: a retrospective analysis of maternal and neonatal outcomes. J Matern Fetal Neonatal Med 2024; 37:2413855. [PMID: 39396881 DOI: 10.1080/14767058.2024.2413855] [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: 07/12/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND This study investigates the effects of administering intravenous esketamine at a dose of 0.25 mg/kg to pregnant patients receiving epidural anesthesia for emergency cesarean section on both maternal and neonatal outcomes. METHODS Medical records of pregnant patients transitioning from labor analgesia to epidural anesthesia for emergency cesarean sections between January 2020 and December 2022 were analyzed. The patients were categorized based on whether they received esketamine infusions during the incision-to-delivery interval. The variables compared between the groups included hemodynamic parameters, perioperative and postoperative adverse reactions, and neonatal outcomes (gender, weight, Apgar scores at 1 and 5 min, need for neonatal intensive care, and umbilical artery/vein blood gas analysis). RESULTS For maternal outcomes, the systolic blood pressure (SBP) in the esketamine group showed a significant increase at 5 and 10 min' post-administration, and the diastolic blood pressure (DBP) significantly increased at 5 min, compared to the control group (p < 0.01). No significant differences were observed in heart rate (HR) and oxygen saturation (SpO2) at any time point (p > 0.05). The esketamine group experienced a significant rise in the incidence of arrhythmias, dizziness, and nystagmus during the perioperative period, a notable decrease in hypotension incidence, and an increase in postoperative nausea and dizziness. Regarding neonatal outcomes, there were no significant differences in gender, weight, Apgar scores ≤7 at 1 and 5 min, and the need for neonatal intensive care. However, the pH level in the umbilical artery blood of the esketamine group was significantly higher. The levels of PCO2 and PO2 in umbilical artery and venous blood did not show significant differences between the groups. CONCLUSIONS In pregnant women undergoing emergency cesarean section, intravenous administration of 0.25 mg/kg esketamine is correlated with favorable maternal and neonatal outcomes.
Collapse
Affiliation(s)
- Xiao-Mei Huang
- Department of Anesthesia, Nantong Maternal and Child Health Hospital, Nantong, China
| | - Hong-Xia Qiu
- Department of Anesthesia, Nantong Maternal and Child Health Hospital, Nantong, China
| |
Collapse
|
2
|
Chae MS, Kim JY, Koh HJ. Early Cognitive Function after Deep Sedation Using Different Anesthetic Agents in Pediatric Patients: A Prospective, Randomized Controlled Trial. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1342. [PMID: 39202623 PMCID: PMC11356384 DOI: 10.3390/medicina60081342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: The impact of anesthetic agents on memory and cognitive function following general anesthesia is of great interest, particularly regarding their effects on the developing pediatric brain. While numerous studies have examined the relationship between anesthetic drugs and brain function, research focusing on early cognitive function following sedation remains limited. Materials and Methods: This study was a prospective, randomized controlled trial involving 148 pediatric patients scheduled for hematological procedures, specifically bone marrow aspiration (BMA) and intrathecal chemotherapy (ITC). Patients were divided into two groups based on the primary anesthetic used: the inhalational sedation group (IHG), in which sevoflurane was used, and the intravenous sedation group (IVG), which received propofol infusion. Apart from the main anesthetic agent, all sedation methods were consistent across both groups. A cognitive function test administered before sedation involved memorizing four distinct images, each associated with a different number. Then, the patients were asked to identify the omitted image upon awakening in the recovery room. Herein, this pre- vs. post-sedation test is called the early recognition assessment (ERA) tool. The primary outcome was the correct response rate after sedation for the two groups. Secondary outcomes included the sedation score, the behavior response score, and the correct response rates according to the number of sedation procedures. Results: This study included 130 patients in the final analysis, with 74 originally assigned to each group. The initial cognitive assessment revealed no significant difference in performance between the anesthetic agents. In addition, no differences were observed in the rates of correct responses or post-sedation scores after repeated procedures. However, the IVG demonstrated higher behavior response scores compared to the IHG. Conclusions: There were no significant differences in the rates of correct responses using the ERA tool between the two groups, irrespective of the number of sedation procedures performed. While some differences were noted in preoperative, intraoperative, and post-anesthesia care, these did not significantly impact the cognitive outcomes measured.
Collapse
Affiliation(s)
| | | | - Hyun Jung Koh
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (M.S.C.); (J.Y.K.)
| |
Collapse
|
3
|
Juneja K, Afroze S, Goti Z, Sahu S, Asawa S, Bhuchakra HP, Natarajan B. Beyond therapeutic potential: a systematic investigation of ketamine misuse in patients with depressive disorders. DISCOVER MENTAL HEALTH 2024; 4:23. [PMID: 38951348 PMCID: PMC11217219 DOI: 10.1007/s44192-024-00077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
Ketamine, a pharmacological agent that acts as an antagonist of the N-methyl-D-aspartate (NMDA) receptor, has garnered considerable interest because of its notable and expeditious antidepressant properties observed in individuals diagnosed with major depressive disorder (MDD) who exhibit resistance to conventional therapeutic interventions. A comprehensive and rigorous systematic review was undertaken to evaluate the prevalence of ketamine abuse undergoing ketamine treatment for depressive disorders. A comprehensive search was conducted across the electronic databases to identify pertinent studies published between 2021 and 2023. The present investigation incorporated a comprehensive range of studies encompassing the abuse or misuse of ketamine, including case reports, observational studies, and clinical trials. Data extraction and quality assessment were conducted in accordance with predetermined criteria. The findings of this systematic review demonstrate the importance of monitoring and addressing ketamine abuse in patients receiving ketamine treatment for depressive disorders like MDD. The wide range of reported prevalence rates highlights the need for standardized criteria and measures for defining and assessing ketamine abuse. This study presents a significant contribution to the field by introducing a novel screening questionnaire and assessment algorithm designed to identify and evaluate ketamine misuse among major depressive disorder (MDD) patients undergoing ketamine treatment. This innovative tool holds the potential to enhance clinical practice by providing healthcare professionals with a standardized approach to promptly detect and address ketamine misuse. The integration of this screening tool into routine care protocols can facilitate more effective monitoring and management of ketamine misuse in this population, ultimately leading to improved patient outcomes and safety.
Collapse
Affiliation(s)
| | - Sabah Afroze
- Shadan Hospital and Institute of Medical Sciences, Hyderabad, India
| | - Zeel Goti
- Government Medical College, Surat, India
| | | | | | | | | |
Collapse
|
4
|
AlGhamdi K, Sadler K. The Use of Ketamine for Malignant and Nonmalignant Chronic Pain in Children: A Review of Current Evidence. J Pain Palliat Care Pharmacother 2024; 38:45-55. [PMID: 38010998 DOI: 10.1080/15360288.2023.2284976] [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: 06/09/2023] [Accepted: 11/05/2023] [Indexed: 11/29/2023]
Abstract
Chronic pain in children continues to pose significant challenges. The pharmacological approach most often revolves around trials and errors, expert opinions, and extrapolation of adult study findings. Ketamine is one of the agents used for chronic pain, especially with a neuropathic component. This article aims to provide an overview of its properties and highlight the current evidence for its use in malignant and nonmalignant chronic pain management. A search on the use of ketamine for chronic pain in children up to 18 years of age covering the period from January 1, 2000, to December 14, 2022, was performed through PubMed, Cochrane Library, EBSCO, EBM Review, Wiley, BMJ, Web of Science, Google Scholar, and the Saudi Digital Library. 218 articles were found and 42 underwent full review. Currently, the evidence about ketamine efficacity and safety for chronic pain management is at best of moderate to low quality. The heterogeinity of ketamine infusion protocols and frequent concomitant use of other analgesics make it difficult to draw robust conclusions. The long-term effect of prolonged usage also remains a concern. Nevertheless, with careful monitoring, the drug may be a reasonable choice for malignant and nonmalignant pain management in selected cases, especially for refractory pain not responding to conventional approaches.
Collapse
Affiliation(s)
- Khaled AlGhamdi
- Consultant Pediatric Complex/Palliative Care, Pediatrics Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Assistant Professor, Al-Faisal University, Riyadh, Saudi Arabia
| | - Kim Sadler
- Advanced Clinical Specialist Nurse, Palliative Care, Oncology Nursing Department, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Loi MV, Lee JH, Huh JW, Mallory P, Napolitano N, Shults J, Krawiec C, Shenoi A, Polikoff L, Al-Subu A, Sanders R, Toal M, Branca A, Glater-Welt L, Ducharme-Crevier L, Breuer R, Parsons S, Harwayne-Gidansky I, Kelly S, Motomura M, Gladen K, Pinto M, Giuliano J, Bysani G, Berkenbosch J, Biagas K, Rehder K, Kasagi M, Lee A, Jung P, Shetty R, Nadkarni V, Nishisaki A. Ketamine Use in the Intubation of Critically Ill Children with Neurological Indications: A Multicenter Retrospective Analysis. Neurocrit Care 2024; 40:205-214. [PMID: 37160847 DOI: 10.1007/s12028-023-01734-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/10/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Ketamine has traditionally been avoided for tracheal intubations (TIs) in patients with acute neurological conditions. We evaluate its current usage pattern in these patients and any associated adverse events. METHODS We conducted a retrospective observational cohort study of critically ill children undergoing TI for neurological indications in 53 international pediatric intensive care units and emergency departments. We screened all intubations from 2014 to 2020 entered into the multicenter National Emergency Airway Registry for Children (NEAR4KIDS) registry database. Patients were included if they were under the age of 18 years and underwent TI for a primary neurological indication. Usage patterns and reported periprocedural composite adverse outcomes (hypoxemia < 80%, hypotension/hypertension, cardiac arrest, and dysrhythmia) were noted. RESULTS Of 21,562 TIs, 2,073 (9.6%) were performed for a primary neurological indication, including 190 for traumatic brain injury/trauma. Patients received ketamine in 495 TIs (23.9%), which increased from 10% in 2014 to 41% in 2020 (p < 0.001). Ketamine use was associated with a coindication of respiratory failure, difficult airway history, and use of vagolytic agents, apneic oxygenation, and video laryngoscopy. Composite adverse outcomes were reported in 289 (13.9%) Tis and were more common in the ketamine group (17.0% vs. 13.0%, p = 0.026). After adjusting for location, patient age and codiagnoses, the presence of respiratory failure and shock, difficult airway history, provider demographics, intubating device, and the use of apneic oxygenation, vagolytic agents, and neuromuscular blockade, ketamine use was not significantly associated with increased composite adverse outcomes (adjusted odds ratio 1.34, 95% confidence interval CI 0.99-1.81, p = 0.057). This paucity of association remained even when only neurotrauma intubations were considered (10.6% vs. 7.7%, p = 0.528). CONCLUSIONS This retrospective cohort study did not demonstrate an association between procedural ketamine use and increased risk of peri-intubation hypoxemia and hemodynamic instability in patients intubated for neurological indications.
Collapse
Affiliation(s)
- Mervin V Loi
- Department of Pediatric Subspecialties, Children's Intensive Care Unit, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, Singapore.
| | - Jan Hau Lee
- Department of Pediatric Subspecialties, Children's Intensive Care Unit, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore, Singapore
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Palen Mallory
- Department of Pediatric Critical Care Medicine, Duke Children's Hospital and Health Center, Durham, NC, USA
| | - Natalie Napolitano
- Respiratory Therapy Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Justine Shults
- Department of Biostatistics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Conrad Krawiec
- Departments of Pediatric Critical Care Medicine and Pediatrics, Penn State Health Children's Hospital, Hershey, PA, USA
| | - Asha Shenoi
- Department of Pediatrics, University of Kentucky, Lexington, KY, USA
| | - Lee Polikoff
- Department of Pediatric Critical Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Awni Al-Subu
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| | - Ronald Sanders
- Division of Critical Care Medicine, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Megan Toal
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Aline Branca
- Department of Pediatric Critical Care Medicine, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Lily Glater-Welt
- Department of Pediatric Critical Care Medicine, Cohen Children's Medical Center of New York, New Hyde Park, NY, USA
| | - Laurence Ducharme-Crevier
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Ryan Breuer
- Division of Critical Care Medicine, Department of Pediatrics, John R. Oishei Children's Hospital, Buffalo, NY, USA
| | - Simon Parsons
- Section of Critical Care Medicine, Alberta Children's Hospital, Calgary, Canada
| | - Ilana Harwayne-Gidansky
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Albany Medical College, Albany, NY, USA
| | - Serena Kelly
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Oregon Health and Science University Doernbecher Children's Hospital, Portland, OR, USA
| | - Makoto Motomura
- Division of Pediatric Critical Care Medicine, Aichi Children's Health and Medical Center, Aichi, Japan
| | - Kelsey Gladen
- Department of Pediatric Critical Care Medicine, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Matthew Pinto
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - John Giuliano
- Section of Pediatric Critical Care, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Gokul Bysani
- Department of Pediatrics, Medical City Children's Hospital, Dallas, TX, USA
| | - John Berkenbosch
- Department of Pediatric Critical Care, University of Louisville and Norton Children's Hospital, Louisville, KY, USA
| | - Katherine Biagas
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Renaissance School of Medicine at Stony, Brook University, Stony Brook, NY, USA
| | - Kyle Rehder
- Division of Pediatric Critical Care, Duke Children's Hospital, Durham, NC, USA
| | - Mioko Kasagi
- Division of Pediatric Critical Care Medicine, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Anthony Lee
- Division of Critical Care Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Philipp Jung
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Rakshay Shetty
- Pediatric Intensive Care, Rainbow Children's Hospital, Bengaluru, India
| | - Vinay Nadkarni
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Akira Nishisaki
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
6
|
Pessano S, Romantsik O, Olsson E, Hedayati E, Bruschettini M. Pharmacological interventions for the management of pain and discomfort during lumbar puncture in newborn infants. Cochrane Database Syst Rev 2023; 9:CD015594. [PMID: 37767875 PMCID: PMC10535798 DOI: 10.1002/14651858.cd015594.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
BACKGROUND Lumbar puncture (LP) is a common invasive procedure, most frequently performed to diagnose infection. Physicians perform LP in newborn infants with the help of an assistant using a strict aseptic technique; it is important to monitor the infant during all the steps of the procedure. Without adequate analgesia, LP can cause considerable pain and discomfort. As newborns have increased sensitivity to pain, it is crucial to adequately manage the procedural pain of LP in this population. OBJECTIVES To assess the benefits and harms, including pain, discomfort, and success rate, of any pharmacological intervention during lumbar puncture in newborn infants, compared to placebo, no intervention, non-pharmacological interventions, or other pharmacological interventions. SEARCH METHODS We searched CENTRAL, PubMed, Embase, and three trial registries in December 2022. We also screened the reference lists of included studies and related systematic reviews for studies not identified by the database searches. SELECTION CRITERIA We included randomized controlled trials (RCTs) and quasi-RCTs comparing drugs used for pain management, sedation, or both, during LP. We considered the following drugs suitable for inclusion. • Topical anesthetics (e.g. eutectic mixture of local anesthetics [EMLA], lidocaine) • Opioids (e.g. morphine, fentanyl) • Alpha-2 agonists (e.g. clonidine, dexmedetomidine) • N-Methyl-D-aspartate (NMDA) receptor antagonists (e.g. ketamine) • Other analgesics (e.g. paracetamol) • Sedatives (e.g. benzodiazepines such as midazolam) DATA COLLECTION AND ANALYSIS: We used standard Cochrane methods. We used the fixed-effect model with risk ratio (RR) for dichotomous data and mean difference (MD) or standardized mean difference (SMD) for continuous data, with their 95% confidence intervals (CIs). Our main outcomes were successful LP on first attempt, total number of LP attempts, episodes of bradycardia, pain assessed with validated scales, episodes of desaturation, number of episodes of apnea, and number of infants with one or more episodes of apnea. We used the GRADE approach to evaluate the certainty of the evidence. MAIN RESULTS We included three studies (two RCTs and one quasi-RCT) that enrolled 206 newborns. One study included only term infants. All studies assessed topical treatment versus placebo or no intervention. The topical anesthetics were lidocaine 4%, lidocaine 1%, and EMLA. We identified no completed studies on opioids, non-steroidal anti-inflammatory drugs, alpha-2 agonists, NMDA receptor antagonists, other analgesics, sedatives, or head-to-head comparisons (drug A versus drug B). Based on very low-certainty evidence from one quasi-RCT of 100 LPs in 76 infants, we are unsure if topical anesthetics (lidocaine), compared to no anesthesia, has an effect on the following outcomes. • Successful LP on first attempt (first-attempts success in 48% of LPs in the lidocaine group and 42% of LPs in the control group) • Number of attempts per LP (mean 1.9 attempts, [standard error of the mean 0.2] in the lidocaine group, and mean 2.1 attempts [standard error of the mean 2.1] in the control group) • Episodes of bradycardia (0% of LPs in the lidocaine group and 4% of LPs in the control group) • Episodes of desaturation (0% of LPs in the lidocaine group and 8% of LPs in the control group) • Occurrence of apnea (RR 3.24, 95% CI 0.14 to 77.79; risk difference [RD] 0.02, 95% CI -0.03 to 0.08). Topical anesthetics compared to placebo may reduce pain assessed with the Neonatal Facial Coding System (NFCS) score (SMD -1.00 standard deviation (SD), 95% CI -1.47 to -0.53; I² = 98%; 2 RCTs, 112 infants; low-certainty evidence). No studies in this comparison reported total number of episodes of apnea. We identified three ongoing studies, which will assess the effects of EMLA, lidocaine, and fentanyl. Three studies are awaiting classification. AUTHORS' CONCLUSIONS The evidence is very uncertain about the effect of topical anesthetics (lidocaine) compared to no anesthesia on successful lumbar puncture on first attempt, the number of attempts per lumbar puncture, episodes of bradycardia, episodes of desaturation, and occurrence of apnea. Compared to placebo, topical anesthetics (lidocaine or EMLA) may reduce pain assessed with the NFCS score. One ongoing study will assess the effects of systemic treatment.
Collapse
Affiliation(s)
- Sara Pessano
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Olga Romantsik
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Faculty of Medicine and Health, School of Health Sciences, Örebro University, Örebro, Sweden
| | - Ehsan Hedayati
- Nezam Mafi Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Matteo Bruschettini
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research and Education, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
7
|
Du Z, Zang Z, Luo J, Liu T, Yang L, Cai Y, Wang L, Zhang D, Zhao J, Gao J, Lv K, Wang L, Li H, Gong H, Fan X. Chronic exposure to (2 R,6 R)-hydroxynorketamine induces developmental neurotoxicity in hESC-derived cerebral organoids. JOURNAL OF HAZARDOUS MATERIALS 2023; 453:131379. [PMID: 37054645 DOI: 10.1016/j.jhazmat.2023.131379] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/25/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023]
Abstract
(R,S)-ketamine (ketamine) has been increasingly used recreationally and medicinally worldwide; however, it cannot be removed by conventional wastewater treatment plants. Both ketamine and its metabolite norketamine have been frequently detected to a significant degree in effluents, aquatic, and even atmospheric environments, which may pose risks to organisms and humans via drinking water and aerosols. Ketamine has been shown to affect the brain development of unborn babies, while it is still elusive whether (2 R,6 R)-hydroxynorketamine (HNK) induces similar neurotoxicity. Here, we investigated the neurotoxic effect of (2 R,6 R)-HNK exposure at the early stages of gestation by applying human cerebral organoids derived from human embryonic stem cells (hESCs). Short-term (2 R,6 R)-HNK exposure did not significantly affect the development of cerebral organoids, but chronic high-concentration (2 R,6 R)-HNK exposure at day 16 inhibited the expansion of organoids by suppressing the proliferation and augmentation of neural precursor cells (NPCs). Notably, the division mode of apical radial glia was unexpectedly switched from vertical to horizontal division planes following chronic (2 R,6 R)-HNK exposure in cerebral organoids. Chronic (2 R,6 R)-HNK exposure at day 44 mainly inhibited the differentiation but not the proliferation of NPCs. Overall, our findings indicate that (2 R,6 R)-HNK administration leads to the abnormal development of cortical organoids, which may be mediated by inhibiting HDAC2. Future clinical studies are needed to explore the neurotoxic effects of (2 R,6 R)-HNK on the early development of the human brain.
Collapse
Affiliation(s)
- Zhulin Du
- School of Life Sciences, Chongqing University, Chongqing, China, Chongqing 401331, China; Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Zhenle Zang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Jing Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Ling Yang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Yun Cai
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Liuyongwei Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Dandan Zhang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Jinghui Zhao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Junwei Gao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Keyi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China
| | - Hong Li
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 40037, China
| | - Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing 40038, China.
| |
Collapse
|
8
|
Yang B, Lian C, Tian R, Chen Y, Tang S, Xiang H, He H, Zhang G. Twelve-year outcomes of bedside laser photocoagulation for severe retinopathy of prematurity. Front Pediatr 2023; 11:1189236. [PMID: 37425279 PMCID: PMC10327642 DOI: 10.3389/fped.2023.1189236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Purpose The purpose of this study is to evaluate the 12-year outcomes of bedside laser photocoagulation (LP) for severe retinopathy of prematurity (ROP) under sedation combined with ocular surface anesthesia in neonatal intensive care units (NICU). Design The study is a retrospective case series. Methods Infants treated with bedside LP for severe ROP from April 2009 to September 2021 were included. All LP treatments were performed under sedation and surface anesthesia at the bedside in NICU. Data were recorded for clinical and demographic characteristics, total laser spots, duration of treatment, proportion of total regression of ROP, proportion of recurrence, and adverse events. Results A total of 364 infants (715 eyes) were included, with a mean gestational age of 28.6 ± 2.4 weeks (range: 22.6-36.6 weeks) and a mean birth weight of 1,156.0 ± 339.0 g (range: 480-2,200 g). The mean number of laser spots was 832 ± 469, and the mean duration of treatment was 23.5 ± 5.3 min per eye. Of all the eyes, 98.3% responded to LP with complete regression of ROP. ROP recurred in 15 (2.1%) eyes after the initial LP. Additional LP was performed in seven (1.0%) eyes. No patient exhibited mistaken LP of other ocular tissues, and there were no serious ocular adverse effects. None of them needed endotracheal intubation. Conclusions Bedside LP treatment is effective and safe for premature infants with severe ROP under sedation and surface anesthesia in NICU, especially for infants whose general condition is unstable and not suitable for transport.
Collapse
Affiliation(s)
- Bingzhi Yang
- Department of Eye Care, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Chaohui Lian
- Department of Eye Care, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Ruyin Tian
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Yi Chen
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Song Tang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Haishan Xiang
- Department of Eye Care, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Honghui He
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Guoming Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| |
Collapse
|
9
|
Xu LL, Wang C, Deng CM, Dai SB, Zhou Q, Peng YB, Shou HY, Han YQ, Yu J, Liu CH, Xia F, Zhang SQ, Wang DX, Chen XZ. Efficacy and Safety of Esketamine for Supplemental Analgesia During Elective Cesarean Delivery: A Randomized Clinical Trial. JAMA Netw Open 2023; 6:e239321. [PMID: 37083664 PMCID: PMC10122167 DOI: 10.1001/jamanetworkopen.2023.9321] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Importance Epidural anesthesia is a primary choice for cesarean delivery, but supplemental analgesics are often required to relieve pain during uterine traction. Objective To investigate the sedative and analgesic effects of intravenous esketamine administered before childbirth via cesarean delivery with the patient under epidural anesthesia. Design, Setting, and Participants This multicenter, double-blind randomized clinical trial assessed 903 women 18 years or older who had full-term single pregnancy and were scheduled for elective cesarean delivery with epidural anesthesia in 5 medical centers in China from September 18, 2021, to September 20, 2022. Intervention Patients were randomized to receive intravenous injection of 0.25 mg/kg of esketamine or placebo before incision. Main Outcomes and Measures The coprimary outcomes included scores on the numeric rating scale of pain (an 11-point scale, with 0 indicating no pain and 10 indicating the worst pain; a difference of ≥1.65 points was clinically meaningful) and Ramsay Sedation Scale (a 6-point scale, with 1 indicating restlessness and 6 indicating deep sleep without response; a difference of ≥2 points was clinically meaningful) immediately after fetal delivery. Secondary outcomes included neonatal Apgar score assessed at 1 and 5 minutes after birth. Results A total of 600 women (mean [SD] age, 30.7 [4.3] years) were enrolled and randomized; all were included in the intention-to-treat analysis. Immediately after fetal delivery, the score on the numeric rating scale of pain was lower with esketamine (median [IQR], 0 [0-1]) than with placebo (median [IQR], 0 [0-2]; median difference, 0; 95% CI, 0-0; P = .001), but the difference was not clinically important. The Ramsay Sedation Scale scores were higher (sedation deeper) with esketamine (median [IQR], 4 [3-4]) than with placebo (median [IQR], 2 [2-2]; median difference, 2; 95% CI, 2-2; P < .001). The neonatal Apgar scores did not differ between the 2 groups at 1 minute (median difference, 0; 95% CI, 0-0; P = .98) and at 5 minutes (median difference, 0; 95% CI, 0-0; P = .27). Transient neurologic or mental symptoms were more common in patients given esketamine (97.7% [293 of 300]) than in those given placebo (4.7% [14 of 300]; P < .001). Conclusions and Relevance For women undergoing cesarean delivery under epidural anesthesia, a subanesthetic dose of esketamine administered before incision produced transient analgesia and sedation but did not induce significant neonatal depression. Mental symptoms and nystagmus were common but transient. Indications and the optimal dose of esketamine in this patient population need further clarification, but study should be limited to those who require supplemental analgesia. Trial Registration ClinicalTrials.gov Identifier: NCT04548973.
Collapse
Affiliation(s)
- Li-Li Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Chun Wang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Chun-Mei Deng
- Department of Anesthesiology and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Shao-Bing Dai
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Qun Zhou
- Department of Anesthesiology, Jiangxi Maternal and Child Health Hospital, Jiangxi Province, China
| | - Yong-Bao Peng
- Department of Anesthesiology, Jiangxi Maternal and Child Health Hospital, Jiangxi Province, China
| | - Hong-Yan Shou
- Department of Anesthesiology, Hangzhou Women's Hospital, Zhejiang, China
| | - Yin-Qiu Han
- Department of Anesthesiology, Hangzhou Women's Hospital, Zhejiang, China
| | - Jing Yu
- Department of Anesthesiology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Chao-Hui Liu
- Department of Anesthesiology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Feng Xia
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang Province, China
| | - Su-Qin Zhang
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, Zhejiang Province, China
| | - Dong-Xin Wang
- Department of Anesthesiology and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Xin-Zhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
10
|
Zhang R, Wang X, Xie Z, Cao T, Jiang S, Huang L. Lipoxin A4 methyl ester attenuated ketamine-induced neurotoxicity in SH-SY5Y cells via regulating leptin pathway. Toxicol In Vitro 2023; 89:105581. [PMID: 36907275 DOI: 10.1016/j.tiv.2023.105581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/18/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023]
Abstract
Ketamine, the widely used intravenous anesthetic, has been reported to cause neurotoxicity and disturbs normal neurogenesis. However, the efficacy of current treatment strategies targeting ketamine's neurotoxicity remains limited. Lipoxin A4 methyl ester (LXA4 ME) is relatively stable lipoxin analog, which serves an important role in protecting against early brain injury. The purpose of this study was to investigate the protective effect of LXA4 ME on ketamine-caused cytotoxicity in SH-SY5Y cells, as well as the underlying mechanisms. Cell viability, apoptosis and endoplasmic reticulum stress (ER stress) were detected by adopting experimental techniques including CCK-8 assay, flow cytometry, western blotting and transmission electron microscope. Furthermore, examining the expression of leptin and its receptor (LepRb), we also measured the levels of activation of the leptin signaling pathway. Our results showed that LXA4 ME intervention promoted the cell viability, inhibited cell apoptosis, and reduced the expression of ER stress related protein and morphological changes induced by ketamine. In addition, inhibition of leptin signaling pathway caused by ketamine could be reversed by LXA4 ME. However, as the specific inhibitor of leptin pathway, leptin antagonist triple mutant human recombinant (leptin tA) attenuated the cytoprotective effect of LXA4 ME against ketamine-induced neurotoxicity. In conclusion, our findings demonstrated LXA4 ME could exert a neuroprotective effect on ketamine-induced neuronal injury via activation of the leptin signaling pathway.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China; Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People's Hospital), No. 1166, Dongfanghong West Road, Decheng District, Dezhou City, Shandong Province, China
| | - Xueji Wang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China; Hebei Medical University, No.48, Donggang Road, Shijiazhuang, Hebei, China
| | - Ziyu Xie
- Hebei Medical University, No.48, Donggang Road, Shijiazhuang, Hebei, China
| | - Tianyu Cao
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China
| | - Sufang Jiang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China
| | - Lining Huang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, No 215 Heping west road, Shijiazhuang, Hebei, China.
| |
Collapse
|
11
|
Acevedo J, Mugarura NE, Welter AL, Johnson EM, Siegel JA. The Effects of Acute and Repeated Administration of Ketamine on Memory, Behavior, and Plasma Corticosterone Levels in Female Mice. Neuroscience 2023; 512:99-109. [PMID: 36496189 DOI: 10.1016/j.neuroscience.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Ketamine is an anesthetic drug that has recently been approved for the treatment of treatment-resistant depression. Females are diagnosed with Major Depressive Disorder at higher rates than males, yet most of the pre-clinical research on ketamine has been conducted in male subjects. Additionally, the literature on the acute and long-term behavioral and cognitive effects of ketamine shows conflicting results. It is important to examine the acute and long-term cognitive and behavioral effects of ketamine exposure at lower sub-anesthetic doses, as the recreational use of the drug at higher doses is associated with cognitive and memory impairments. The current study examined the effects of acute and repeated ketamine exposure on anxiety-like behavior, novel object recognition memory, depression-like behavior, and plasma corticosterone levels in 20 adult female C57BL/6J mice. Mice were exposed acutely or repeatedly for 10 consecutive days to saline or 15 mg/kg ketamine and behavior was measured in the open field test, novel object recognition test, and the Porsolt forced swim test. Plasma corticosterone levels were measured following behavioral testing. Acute ketamine exposure decreased locomotor activity and increased anxiety-like behavior in the open field test compared to controls, while repeated ketamine exposure impaired memory in the novel object recognition test. There were no effects of acute or repeated ketamine exposure on depression-like behavior in the Porsolt forced swim test or on plasma corticosterone levels. These findings suggest that a subanesthetic dose of ketamine alters behavior and cognition in female mice and the effects are dependent on the duration of exposure.
Collapse
Affiliation(s)
- Jonathan Acevedo
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W Carson St, Torrance, CA 90502, USA.
| | - Naomi E Mugarura
- Neuroscience Program, University of St. Thomas, 2115 Summit Ave, Saint Paul, MN 55105, USA.
| | - Alex L Welter
- Neuroscience Program, University of St. Thomas, 2115 Summit Ave, Saint Paul, MN 55105, USA.
| | - Emily M Johnson
- Neuroscience Program, University of St. Thomas, 2115 Summit Ave, Saint Paul, MN 55105, USA.
| | - Jessica A Siegel
- Department of Biochemistry and Biophysics, The College of Science, Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331, USA.
| |
Collapse
|
12
|
Dawes JM, Howard RF. Neonatal Pain: Significance, Assessment, and Management. NEONATAL ANESTHESIA 2023:505-527. [DOI: 10.1007/978-3-031-25358-4_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Pessano S, Romantsik O, Hedayati E, Olsson E, Bruschettini M. Pharmacological interventions for the management of pain and discomfort during lumbar puncture in newborn infants. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2022; 2022:CD015594. [PMCID: PMC9749080 DOI: 10.1002/14651858.cd015594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To assess the benefits and harms, including pain, discomfort, and success rate, of any pharmacological intervention during lumbar puncture in newborn infants, compared to placebo, no intervention, non‐pharmacological interventions, or other pharmacological interventions.
Collapse
Affiliation(s)
| | - Sara Pessano
- Neonatal Intensive Care Unit, Department Mother and ChildIRCCS Istituto Giannina GasliniGenoaItaly
| | - Olga Romantsik
- Department of Clinical Sciences Lund, PaediatricsLund University, Skåne University HospitalLundSweden
| | - Ehsan Hedayati
- Nezam Mafi HospitalAhvaz Jundishapur University of Medical SciencesAhvazIran
| | | | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University HospitalLundSweden,Cochrane Sweden, Lund University, Skåne University HospitalLundSweden
| |
Collapse
|
14
|
Acevedo J, Siegel JA. Neurobiological, behavioral, and cognitive effects of ketamine in adolescents: A review of human and pre-clinical research. Behav Brain Res 2022; 435:114049. [PMID: 35952776 DOI: 10.1016/j.bbr.2022.114049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/01/2022] [Accepted: 08/07/2022] [Indexed: 11/29/2022]
Abstract
S-ketamine is approved for treatment-resistant patients with depression and adult patients with suicide behavior. While ketamine is therapeutically beneficial in adults, there is a dearth of research on the effects of ketamine on adolescent brain function and behavior. In this review we summarize the current literature on the neurobiological and behavioral effects of adolescent ketamine exposure in preclinical animal models and humans. A search of PubMed was conducted using pre-defined criteria, resulting in the evaluation of 406 articles. A total of 39 animal studies and 7 human studies met the selection criteria. The included studies examined the effects of ketamine exposure during adolescence and excluded studies on ketamine use for pain or anesthesia and ketamine as a model of schizophrenia. Pre-clinical animal models of adolescent ketamine exposure show ketamine-induced neurotoxicity and apoptosis, and changes in locomotor activity, social behaviors, anxiety- and depression-like behaviors, and memory. There is variability in the results, and differences in ketamine dose and length of exposure appears to influence the results. Ketamine reduces symptoms of depression and anxiety and improves mood in human adolescents. Much of the literature on adolescent ketamine exposure examines the effects in males, with more limited research in females. Relatively little research has focused on adolescent ketamine exposure. Despite its effectiveness for mitigating symptoms of depression, adolescent ketamine exposure can disrupt memory and other behaviors and have deleterious effects on brain function. Further research is warranted to better define doses and dosing paradigms that are beneficial without unintended side effects in adolescence.
Collapse
Affiliation(s)
- Jonathan Acevedo
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W Carson St, Torrance, CA 90502, USA.
| | - Jessica A Siegel
- Department of Biochemistry and Biophysics, The College of Science, Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331, USA.
| |
Collapse
|
15
|
Scutellaria baicalensis and its constituents baicalin and baicalein as antidotes or protective agents against chemical toxicities: a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:1297-1329. [PMID: 35676380 DOI: 10.1007/s00210-022-02258-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/21/2022] [Indexed: 10/18/2022]
Abstract
Scutellaria baicalensis (SB), also known as the Chinese skullcap, has a long history of being used in Chinese medicine to treat a variety of conditions ranging from microbial infections to metabolic syndrome and malignancies. Numerous studies have reported that treatment with total SB extract or two main flavonoids found in its root and leaves, baicalin (BA) and baicalein (BE), can prevent or alleviate the detrimental toxic effects of exposure to various chemical compounds. It has been shown that BA and BE are generally behind the protective effects of SB against toxicants. This paper aimed to review the protective and therapeutic effects of SB and its main components BA and BE against chemical compounds that can cause intoxication after acute or chronic exposure and seriously affect different vital organs including the brain, heart, liver, and kidneys. In this review paper, we had a look into a total of 221 in vitro and in vivo studies from 1995 to 2021 from the scientific databases PubMed, Scopus, and Web of Science which reported protective or therapeutic effects of BA, BE, or SB against drugs and chemicals that one might be exposed to on a professional or accidental basis and compounds that are primarily used to simulate disease models. In conclusion, the protective effects of SB and its flavonoids can be mainly attributed to increase in antioxidants enzymes, inhibition of lipid peroxidation, reduction of inflammatory cytokines, and suppression of apoptosis pathway.
Collapse
|
16
|
Durrmeyer X, Walter-Nicolet E, Chollat C, Chabernaud JL, Barois J, Chary Tardy AC, Berenguer D, Bedu A, Zayat N, Roué JM, Beissel A, Bellanger C, Desenfants A, Boukhris R, Loose A, Massudom Tagny C, Chevallier M, Milesi C, Tauzin M. Premedication before laryngoscopy in neonates: Evidence-based statement from the French society of neonatology (SFN). Front Pediatr 2022; 10:1075184. [PMID: 36683794 PMCID: PMC9846576 DOI: 10.3389/fped.2022.1075184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/01/2022] [Indexed: 01/06/2023] Open
Abstract
CONTEXT Laryngoscopy is frequently required in neonatal intensive care. Awake laryngoscopy has deleterious effects but practice remains heterogeneous regarding premedication use. The goal of this statement was to provide evidence-based good practice guidance for clinicians regarding premedication before tracheal intubation, less invasive surfactant administration (LISA) and laryngeal mask insertion in neonates. METHODS A group of experts brought together by the French Society of Neonatology (SFN) addressed 4 fields related to premedication before upper airway access in neonates: (1) tracheal intubation; (2) less invasive surfactant administration; (3) laryngeal mask insertion; (4) use of atropine for the 3 previous procedures. Evidence was gathered and assessed on predefined questions related to these fields. Consensual statements were issued using the GRADE methodology. RESULTS Among the 15 formalized good practice statements, 2 were strong recommendations to do (Grade 1+) or not to do (Grade 1-), and 4 were discretionary recommendations to do (Grade 2+). For 9 good practice statements, the GRADE method could not be applied, resulting in an expert opinion. For tracheal intubation premedication was considered mandatory except for life-threatening situations (Grade 1+). Recommended premedications were a combination of opioid + muscle blocker (Grade 2+) or propofol in the absence of hemodynamic compromise or hypotension (Grade 2+) while the use of a sole opioid was discouraged (Grade 1-). Statements regarding other molecules before tracheal intubation were expert opinions. For LISA premedication was recommended (Grade 2+) with the use of propofol (Grade 2+). Statements regarding other molecules before LISA were expert opinions. For laryngeal mask insertion and atropine use, no specific data was found and expert opinions were provided. CONCLUSION This statement should help clinical decision regarding premedication before neonatal upper airway access and favor standardization of practices.
Collapse
Affiliation(s)
- Xavier Durrmeyer
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Créteil, Créteil, France.,Université Paris Est Créteil, Faculté de Santé de Créteil, IMRB, GRC CARMAS, Créteil, France
| | - Elizabeth Walter-Nicolet
- Neonatal Medicine and Intensive Care Unit, Saint Joseph Hospital, Paris, France.,University of Paris-Cité, CRESS, Obstetrical Perinatal and Pediatric Epidemiology Research Team, EPOPé, INSERM, INRAE, Paris, France
| | - Clément Chollat
- Department of Neonatology, Hôpital Armand Trousseau, APHP, Sorbonne Université, Paris, France
| | - Jean-Louis Chabernaud
- Division of Neonatal and Pediatric Critical Care Transportation, Hôpital Antoine Beclere, AP-HP, Paris - Saclay University Hospital, Clamart, France
| | - Juliette Barois
- Department of Neonatology and Neonatal Intensive Care, CH de Valenciennes, Valenciennes, France
| | - Anne-Cécile Chary Tardy
- Department of Neonatology and Neonatal Intensive Care, Centre Hospitalier Universitaire de Dijon, Dijon, France
| | - Daniel Berenguer
- Department of Pediatric Anesthesia and Pediatric Transport (SMUR Pédiatrique), Hôpital des Enfants, CHU de Bordeaux, Bordeaux, France
| | - Antoine Bedu
- Department of Neonatal Pediatrics and Intensive Care, Limoges University Hospital, Limoges, France
| | - Noura Zayat
- Department of Neonatal Intensive Care and Pediatric Transport, CHU de Nantes, Nantes, France
| | - Jean-Michel Roué
- Department of Pediatric and Neonatal Critical Care, Brest University Hospital, Brest, France
| | - Anne Beissel
- Neonatal Intensive Care Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Claire Bellanger
- Department of Neonatology and Neonatal Intensive Care, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| | - Aurélie Desenfants
- Department of Neonatology, CHU Nimes, Université Montpellier, Nimes, France
| | - Riadh Boukhris
- Department of Neonatology, Pôle Femme-Mère-Nouveau-Né, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Anne Loose
- Department of Neonatology, CHRU de Tours, Hôpital Bretonneau, Tours, France
| | - Clarisse Massudom Tagny
- Department of Neonatology and Neonatal Intensive Care, Grand Hôpital de L'Est Francilien, Meaux, France
| | - Marie Chevallier
- Department of Neonatal Intensive Care Unit, CHU Grenoble, Grenoble, France.,TIMC-IMAG Research Department, Grenoble Alps University, Grenoble, France
| | - Christophe Milesi
- Department of Neonatal Medicine and Pediatric Intensive Care, Montpellier University Hospital, Université de Montpellier, Montpellier, France
| | - Manon Tauzin
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| |
Collapse
|
17
|
Song B, Zhu JC. Mechanisms of the Rapid Effects of Ketamine on Depression and Sleep Disturbances: A Narrative Review. Front Pharmacol 2022; 12:782457. [PMID: 34970147 PMCID: PMC8712478 DOI: 10.3389/fphar.2021.782457] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/22/2021] [Indexed: 12/24/2022] Open
Abstract
Recently, sleep has been recognized as a crucial factor for health and longevity. The daily sleep/wake cycle provides the basis of biorhythm, which controls whole-body homeostasis and homeodynamics. Sleep disturbances can contribute to several physical and psychological disorders, including cardiovascular disease, obesity, depression, and cognitive dysfunction. The clinical use of the N-methyl-D-aspartate (NMDA) receptor antagonist ketamine began in the 1970s. Over the years, physicians have used it as a short-acting anesthetic, analgesic, and antidepressant; however, in-depth research has revealed new possible applications for ketamine, such as for treating sleep disturbances and circadian rhythm disorders. The aim of this narrative review is to examine the literature on the mechanistic role of the antidepressant ketamine in affecting sleep disturbance. Additionally, we discuss the pharmacologic and pharmacokinetic mechanisms of ketamine as an antidepressant and the predictive biomarkers for ketamine’s effect on sleep and cognitive function.
Collapse
Affiliation(s)
- Bijia Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun-Chao Zhu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Choudhury D, Autry AE, Tolias KF, Krishnan V. Ketamine: Neuroprotective or Neurotoxic? Front Neurosci 2021; 15:672526. [PMID: 34566558 PMCID: PMC8461018 DOI: 10.3389/fnins.2021.672526] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022] Open
Abstract
Ketamine, a non-competitive N-methyl-D-aspartate receptor (NMDAR) antagonist, has been employed clinically as an intravenous anesthetic since the 1970s. More recently, ketamine has received attention for its rapid antidepressant effects and is actively being explored as a treatment for a wide range of neuropsychiatric syndromes. In model systems, ketamine appears to display a combination of neurotoxic and neuroprotective properties that are context dependent. At anesthetic doses applied during neurodevelopmental windows, ketamine contributes to inflammation, autophagy, apoptosis, and enhances levels of reactive oxygen species. At the same time, subanesthetic dose ketamine is a powerful activator of multiple parallel neurotrophic signaling cascades with neuroprotective actions that are not always NMDAR-dependent. Here, we summarize results from an array of preclinical studies that highlight a complex landscape of intracellular signaling pathways modulated by ketamine and juxtapose the somewhat contrasting neuroprotective and neurotoxic features of this drug.
Collapse
Affiliation(s)
- Divya Choudhury
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Anita E. Autry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Vaishnav Krishnan
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Department of Neurology, Baylor College of Medicine, Houston, TX, United States
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
19
|
Kirmani BF, Au K, Ayari L, John M, Shetty P, Delorenzo RJ. Super-Refractory Status Epilepticus: Prognosis and Recent Advances in Management. Aging Dis 2021; 12:1097-1119. [PMID: 34221552 PMCID: PMC8219503 DOI: 10.14336/ad.2021.0302] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Super-refractory status epilepticus (SRSE) is a life-threatening neurological emergency with high morbidity and mortality. It is defined as “status epilepticus (SE) that continues or recurs 24 hours or more after the onset of anesthesia, including those cases in which SE recurs on the reduction or withdrawal of anesthesia.” This condition is resistant to normal protocols used in the treatment of status epilepticus and exposes patients to increased risks of neuronal death, neuronal injury, and disruption of neuronal networks if not treated in a timely manner. It is mainly seen in patients with severe acute onset brain injury or presentation of new-onset refractory status epilepticus (NORSE). The mortality, neurological deficits, and functional impairments are significant depending on the duration of status epilepticus and the resultant brain damage. Research is underway to find the cure for this devastating neurological condition. In this review, we will discuss the wide range of therapies used in the management of SRSE, provide suggestions regarding its treatment, and comment on future directions. The therapies evaluated include traditional and alternative anesthetic agents with antiepileptic agents. The other emerging therapies include hypothermia, steroids, immunosuppressive agents, electrical and magnetic stimulation therapies, emergent respective epilepsy surgery, the ketogenic diet, pyridoxine infusion, cerebrospinal fluid drainage, and magnesium infusion. To date, there is a lack of robust published data regarding the safety and effectiveness of various therapies, and there continues to be a need for large randomized multicenter trials comparing newer therapies to treat this refractory condition.
Collapse
Affiliation(s)
- Batool F Kirmani
- 1Texas A&M University College of Medicine, College Station, TX, USA.,3Epilepsy and Functional Neurosurgery Program, Department of Neurology, CHI St. Joseph Health, Bryan, TX, USA
| | - Katherine Au
- 2George Washington University, School of Medicine & Health Sciences, Washington DC, USA
| | - Lena Ayari
- 1Texas A&M University College of Medicine, College Station, TX, USA
| | - Marita John
- 1Texas A&M University College of Medicine, College Station, TX, USA
| | - Padmashri Shetty
- 4M. S. Ramaiah Medical College, M. S. Ramaiah Nagar, Bengaluru, Karnataka, India
| | - Robert J Delorenzo
- 5Department of Neurology, Virginia Commonwealth University School of Medicine, Richmond, VA
| |
Collapse
|
20
|
Zhang Z, Liu W, Shen M, Ma X, Li R, Jin X, Bai H, Gao L. Protective Effect of GM1 Attenuates Hippocampus and Cortex Apoptosis After Ketamine Exposure in Neonatal Rat via PI3K/AKT/GSK3β Pathway. Mol Neurobiol 2021; 58:3471-3483. [PMID: 33733293 DOI: 10.1007/s12035-021-02346-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/01/2021] [Indexed: 12/21/2022]
Abstract
Ketamine is a widely used analgesic and anesthetic in obstetrics and pediatrics. Ketamine is known to promote neuronal death and cognitive dysfunction in the brains of humans and animals during development. Monosialotetrahexosyl ganglioside (GM1), a promoter of brain development, exerts neuroprotective effects in many neurological disease models. Here, we investigated the neuroprotective effect of GM1 and its potential underlying mechanism against ketamine-induced apoptosis of rats. Seven-day-old Sprague Dawley (SD) rats were randomly divided into the following four groups: (1) group C (control group: normal saline was injected intraperitoneally); (2) group K (ketamine); (3) group GM1 (GM1 was given before normal saline injection); and (4) GM1+K group (received GM1 30 min before continuous exposure to ketamine). Each group contained 15 rats, received six doses of ketamine (20 mg/kg), and was injected with saline every 90 min. The Morris water maze (MWM) test, the number of cortical and hippocampal cells, apoptosis, and AKT/GSK3β pathway were analyzed. To determine whether GM1 exerted its effect via the PI3K/AKT/GSK3β pathway, PC12 cells were incubated with LY294002, a PI3K inhibitor. We found that GM1 protected against ketamine-induced apoptosis in the hippocampus and cortex by reducing the expression of Bcl-2 and Caspase-3, and by increasing the expression of Bax. GM1 treatment increased the expression of p-AKT and p-GSK3β. However, the anti-apoptotic effect of GM1 was eliminated after inhibiting the phosphorylation of AKT. We showed that GM1 lessens ketamine-induced apoptosis in the hippocampus and cortex of young rats by regulating the PI3K/AKT/GSK3β pathway. Taken together, GM1 may be a potential preventive treatment for the neurotoxicity caused by continuous exposure to ketamine.
Collapse
Affiliation(s)
- Zhiheng Zhang
- College of Veterinary Medicine, Northeast Agricultural University, No. 600 Changjiang Rd, Xiangfang District, Harbin, 150030, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agriculture University, Harbin, China
| | - Wenhan Liu
- School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Meilun Shen
- College of Veterinary Medicine, Northeast Agricultural University, No. 600 Changjiang Rd, Xiangfang District, Harbin, 150030, China
| | - Xiangying Ma
- College of Veterinary Medicine, Northeast Agricultural University, No. 600 Changjiang Rd, Xiangfang District, Harbin, 150030, China
| | - Rouqian Li
- College of Veterinary Medicine, Northeast Agricultural University, No. 600 Changjiang Rd, Xiangfang District, Harbin, 150030, China
| | - Xiaodi Jin
- College of Veterinary Medicine, Northeast Agricultural University, No. 600 Changjiang Rd, Xiangfang District, Harbin, 150030, China
| | - Hui Bai
- College of Veterinary Medicine, Northeast Agricultural University, No. 600 Changjiang Rd, Xiangfang District, Harbin, 150030, China
| | - Li Gao
- College of Veterinary Medicine, Northeast Agricultural University, No. 600 Changjiang Rd, Xiangfang District, Harbin, 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agriculture University, Harbin, China.
| |
Collapse
|
21
|
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
|
22
|
Young JT, Vlasova RM, Howell BR, Knickmeyer RC, Morin E, Kuitchoua KI, Lubach GR, Noel J, Hu X, Shi Y, Caudill G, Alexander AL, Niethammer M, Paule MG, Coe CL, Sanchez M, Styner M. General anaesthesia during infancy reduces white matter micro-organisation in developing rhesus monkeys. Br J Anaesth 2021; 126:845-853. [PMID: 33549320 DOI: 10.1016/j.bja.2020.12.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 12/04/2020] [Accepted: 12/24/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Non-human primates are commonly used in neuroimaging research for which general anaesthesia or sedation is typically required for data acquisition. In this analysis, the cumulative effects of exposure to ketamine, Telazol® (tiletamine and zolazepam), and the inhaled anaesthetic isoflurane on early brain development were evaluated in two independent cohorts of typically developing rhesus macaques. METHODS Diffusion MRI scans were analysed from 43 rhesus macaques (20 females and 23 males) at either 12 or 18 months of age from two separate primate colonies. RESULTS Significant, widespread reductions in fractional anisotropy with corresponding increased axial, mean, and radial diffusivity were observed across the brain as a result of repeated anaesthesia exposures. These effects were dose dependent and remained after accounting for age and sex at time of exposure in a generalised linear model. Decreases of up to 40% in fractional anisotropy were detected in some brain regions. CONCLUSIONS Multiple exposures to commonly used anaesthetics were associated with marked changes in white matter microstructure. This study is amongst the first to examine clinically relevant anaesthesia exposures on the developing primate brain. It will be important to examine if, or to what degree, the maturing brain can recover from these white matter changes.
Collapse
Affiliation(s)
- Jeffrey T Young
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Roza M Vlasova
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Brittany R Howell
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Rebecca C Knickmeyer
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elyse Morin
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kaela I Kuitchoua
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Gabriele R Lubach
- Harlow Center for Biological Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jean Noel
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiaoping Hu
- Department of Bioengineering, University of California, Riverside, CA, USA
| | - Yundi Shi
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gibson Caudill
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew L Alexander
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, USA
| | - Marc Niethammer
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Merle G Paule
- Division of Neurotoxicology, National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR, USA
| | - Christopher L Coe
- Harlow Center for Biological Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Mar Sanchez
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Martin Styner
- Department of Psychiatry University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Wang Z, Han S, Cai M, Du P, Zhang Z, Li X. Environmental behavior of methamphetamine and ketamine in aquatic ecosystem: Degradation, bioaccumulation, distribution, and associated shift in toxicity and bacterial community. WATER RESEARCH 2020; 174:115585. [PMID: 32105996 DOI: 10.1016/j.watres.2020.115585] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/26/2020] [Accepted: 02/01/2020] [Indexed: 06/10/2023]
Abstract
Ketamine (KET) and methamphetamine (METH) have been recognized as emerging contaminants in aquatic ecosystems. This paper aimed to investigate the environmental behaviour, including the degradation, distribution, and bioaccumulation, of METH, KET, and their main metabolites (amphetamine (AMP) and norketamine (NorKET)). The changes in acute toxicity in the aqueous phase and in the bacterial community in sediment were determined to assess the associated eco-risk of the drug exposure. Five types of lab-scale aquatic ecosystems were established and exposed to KET or METH for 40 days: a water- sediment- organisms- KET system (K), a water- sediment- organisms- METH system (M), a water- sediment- organism- METH- KET system (M + K), a water-sediment- KET- METH system (control), and a water- sediment- organisms system (biocontrol). The results demonstrated that much faster degradation occurred for both METH (t1/2 = 3.89 and 2.37 days in the M and M + K group, respectively) and KET(t1/2 = 5.69 days 5.39 days in the K group and M + K group, respectively) than in the control group (t1/2 = 7.83 and 86.71days for METH and KET, respectively). Rapid adsorption of KET, METH, and their metabolites was observed in the sediment, which had clay and silt as the main particle sizes. KET was observed to be absorbed by shallow-water fish (Chinese medaka, rosy bitterling and mosquito fish), while METH was dominantly ingested by bottom-dwellers (loach). Duckweed might play a crucial role in the dissipation process of METH and KET, which were mainly adsorbed by duckweed roots. During incubation, the acute toxic levels in the K and M + K groups changed from non-toxic to medium toxicity levels, and the toxicity in the M and control groups changed from non-toxic to low toxicity levels. Moreover, marked changes in the bacterial community in the sediment induced by METH or KET exposure were observed, and the most significant change in the bacterial community was observed in the group spiked with both METH and KET. This work for the first time elucidated the environmental behaviors of METH and KET in aquatic ecosystem and associated the impact on ecological system equilibrium.
Collapse
Affiliation(s)
- Zhenglu Wang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China; Institute of Marine Biology, College of Oceanography, Hohai University, Nanjing, Jiangsu, 210098, PR China
| | - Sheng Han
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China
| | - Min Cai
- Eco-environmental Protection Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201403, PR China
| | - Peng Du
- Beijing Key Laboratory of Urban Hydrological Cycle and Sponge City Technology, College of Water Sciences, Beijing Normal University, Beijing, 100875, PR China
| | - Zhaobin Zhang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China
| | - Xiqing Li
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China.
| |
Collapse
|
24
|
Pribish A, Wood N, Kalava A. A Review of Nonanesthetic Uses of Ketamine. Anesthesiol Res Pract 2020; 2020:5798285. [PMID: 32308676 PMCID: PMC7152956 DOI: 10.1155/2020/5798285] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/07/2020] [Accepted: 03/05/2020] [Indexed: 12/20/2022] Open
Abstract
Ketamine, a nonselective NMDA receptor antagonist, is used widely in medicine as an anesthetic agent. However, ketamine's mechanisms of action lead to widespread physiological effects, some of which are now coming to the forefront of research for the treatment of diverse medical disorders. This paper aims at reviewing recent data on key nonanesthetic uses of ketamine in the current literature. MEDLINE, CINAHL, and Google Scholar databases were queried to find articles related to ketamine in the treatment of depression, pain syndromes including acute pain, chronic pain, and headache, neurologic applications including neuroprotection and seizures, and alcohol and substance use disorders. It can be concluded that ketamine has a potential role in the treatment of all of these conditions. However, research in this area is still in its early stages, and larger studies are required to evaluate ketamine's efficacy for nonanesthetic purposes in the general population.
Collapse
Affiliation(s)
- Abby Pribish
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nicole Wood
- Department of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Arun Kalava
- Department of Anesthesiology, University of Central Florida College of Medicine, Orlando, FL, USA
| |
Collapse
|
25
|
Li Q, Qiu Z, Lu Y, Lu P, Wen J, Wang K, Zhao X, Li R, Zhang H, Zhang Y, Jia P, Fan P, Zhang Y, Zhang S, Lu H, Chen X, Liu Y, Zhang P. Edaravone protects primary-cultured rat cortical neurons from ketamine-induced apoptosis via reducing oxidative stress and activating PI3K/Akt signal pathway. Mol Cell Neurosci 2019; 100:103399. [DOI: 10.1016/j.mcn.2019.103399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 07/12/2019] [Accepted: 08/25/2019] [Indexed: 12/27/2022] Open
|
26
|
The effects of sub-anesthetic ketamine plus ethanol on behaviors and apoptosis in the prefrontal cortex and hippocampus of adolescent rats. Pharmacol Biochem Behav 2019; 184:172742. [PMID: 31348944 DOI: 10.1016/j.pbb.2019.172742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/10/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022]
Abstract
Ketamine has become increasingly popular in adolescent drug abusers worldwide. Meanwhile, alcohol is usually used by ketamine users. However, little work has been conducted to examine the chronic combined effects of ketamine and ethanol on adolescent brain. Here we probed into the effects of chronic administration of ketamine at recreational doses alone or combined with ethanol on behaviors and neuron damage in an adolescent rat model. 28-day old rats were treated with either 20 or 30 mg/kg ketamine plus or not plus 10% ethanol daily for 21 days. Depressive like behaviors, anxiety like behavior and memory impairment were tested using open field test, forced swimming test, elevated plus maze and Morris water maze. Apoptosis in prefrontal cortex (PFC) and hippocampus (HIP) were determined by the TdT-mediated dUTP Nick-End Labeling (TUNEL) and protein and mRNA levels of caspase-3, Bax and Bcl-2. Results show that co-application of ketamine and ethanol significantly increased immobility time in the forced swimming test, up-regulated TUNEL positive cells and both protein and mRNA expressions of caspase-3 and Bax, compared with the control group and ketamine and ethanol use alone groups in the PFC, but not in the HIP. Our study suggests that chronic co-administration of ketamine and ethanol results in depressive-like behavior and the caspase-dependent apoptosis in the PFC of adolescent rats' brains.
Collapse
|
27
|
Neonatal exposure to the experimental environment or ketamine can induce long-term learning dysfunction or overmyelination in female but not male rats. Neuroreport 2019; 30:491-497. [PMID: 30920435 PMCID: PMC6493779 DOI: 10.1097/wnr.0000000000001228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ketamine can induce neurotoxicity after exposures to the developing brain. To investigate whether ketamine at subanesthetic dosage or its environmental condition can cause long-term cognitive dysfunction after multiple exposures in male or female neonatal rats, postnatal day 5 (P5)-day-old Sprague-Dawley rats were randomized into three groups: ketamine group, vehicle group, and control group (no disturbance). Learning and memory abilities from P60 to P65 and immunofluorescence tests for myelin basic protein (MBP) in gray matter on P65 were conducted. The results showed that in female rats, the path length on day 1 in ketamine group and on days 1 and 2 in vehicle group was longer than that in control (P<0.05), but there was no difference between ketamine and vehicle groups (P>0.05). The mean density of MBP in the medial prefrontal cortex (mPFC) was significantly increased in vehicle and ketamine groups compared with that in control (P<0.05), and there was a significant difference between vehicle and ketamine groups (P<0.05), but MBP density was not changed in CA1 or CA3 region (P>0.05). In male rats, there were no significant differences in path length among the groups, and the density of MBP in the mPFC and hippocampus in vehicle or ketamine group was not different from that in control (P>0.05). Pearson’s correlation analysis showed that there was a positive correlation between MBP density in the mPFC and path length in adult female rats (r=0.753, P<0.01). Overall, the results suggested that neonatal female rats exposed to multiple episodes of the experimental environment can develop learning dysfunction in adulthood, which may result from overmyelination in the mPFC, but male rats were not affected. Ketamine could increase myelination in the mPFC in female rats, but it did not induce learning dysfunction in adulthood; therefore, ketamine may be a safe drug for pediatric anesthesia.
Collapse
|
28
|
Walter-Nicolet E, Courtois E, Milesi C, Ancel PY, Beuchée A, Tourneux P, Benhammou V, Carbajal R, Durrmeyer X. Premedication practices for delivery room intubations in premature infants in France: Results from the EPIPAGE 2 cohort study. PLoS One 2019; 14:e0215150. [PMID: 30970001 PMCID: PMC6457540 DOI: 10.1371/journal.pone.0215150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 03/27/2019] [Indexed: 11/19/2022] Open
Abstract
Objectives To assess premedication practices before tracheal intubation of premature newborns in the delivery room (DR). Study design From the national population-based prospective EPIPAGE 2 cohort in 2011, we extracted all live born preterms intubated in the DR in level-3 centers, without subsequent circulatory resuscitation. Studied outcomes included the rate and type of premedication, infants’ and maternities’ characteristics and survival and major neonatal morbidities at discharge from hospital. Univariate and multivariate analysis were performed and a generalized estimating equation was used to identify factors associated with premedication use. Results Out of 1494 included neonates born in 65 maternities, 76 (5.1%) received a premedication. Midazolam was the most used drug accounting for 49% of the nine drugs regimens observed. Premedicated, as compared to non premedicated neonates, had a higher median [IQR] gestational age (30 [28–31] vs 28 [27–30] weeks, p<10−3), median birth weight (1391 [1037–1767] vs 1074 [840–1440] g, p<10−3) and median 1-minute Apgar score (8 [6–9] vs 6 [3–8], p<10−3). Using univariate analyses, premedication was significantly less frequent after maternal general anesthesia and during nighttime and survival without major morbidity was significantly higher among premedicated neonates (56/73 (81.4%) vs 870/1341 (69.3%), p = 0.028). Only 10 centers used premedication at least once and had characteristics comparable to the 55 other centers. In these 10 centers, premedication rates varied from 2% to 75%, and multivariate analysis identified gestational age and 1-minute Apgar score as independent factors associated with premedication use. Conclusion Premedication rate before tracheal intubation was only 5.1% in the DR of level-3 maternities for premature neonates below 34 weeks of gestation in France in 2011 and seemed to be mainly associated with centers’ local policies.
Collapse
Affiliation(s)
| | - Emilie Courtois
- Paediatric Emergency Department. Trousseau Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Christophe Milesi
- Paediatric and Neonatal Intensive Care Unit, University Hospital Arnaud de Villeneuve, Montpellier, France
| | - Pierre-Yves Ancel
- INSERM, U1153, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Obstetrical, Perinatal and Pediatric Epidemiology Team, Paris, France
- Paris Descartes University France, Paris, France
- URC - CIC P1419, Cochin Hotel-Dieu Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Alain Beuchée
- Division of Neonatology and CIC-1414, Department of Pediatrics, University Hospital, Rennes, France
- LTSI, Inserm U1099, Université de Rennes 1, Rennes, France
| | - Pierre Tourneux
- Neonatal and Paediatric Intensive Care Unit, University hospital, Amiens, France
- PériTox - UMI 01, Medicine University, Picardie Jules Verne University, Amiens, France
| | - Valérie Benhammou
- INSERM, U1153, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Obstetrical, Perinatal and Pediatric Epidemiology Team, Paris, France
- Paris Descartes University France, Paris, France
| | - Ricardo Carbajal
- Paediatric Emergency Department. Trousseau Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
- INSERM, U1153, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Obstetrical, Perinatal and Pediatric Epidemiology Team, Paris, France
- Paris Descartes University France, Paris, France
- Paediatric and Neonatal Intensive Care Unit, Trousseau Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Xavier Durrmeyer
- INSERM, U1153, Epidemiology and Statistics Sorbonne Paris Cité Research Center, Obstetrical, Perinatal and Pediatric Epidemiology Team, Paris, France
- Neonatal Intensive Care Unit, Centre Hospitalier Intercommunal de Créteil, University Paris Est Créteil, Créteil, France
| |
Collapse
|
29
|
Zheng W, Zhou YL, Liu WJ, Wang CY, Zhan YN, Li HQ, Chen LJ, Li MD, Ning YP. Neurocognitive performance and repeated-dose intravenous ketamine in major depressive disorder. J Affect Disord 2019; 246:241-247. [PMID: 30590286 DOI: 10.1016/j.jad.2018.12.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/21/2018] [Accepted: 12/08/2018] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Ketamine has demonstrated a rapid antidepressant and antisuicidal effect in patients with major depressive disorder (MDD), but the neurocognitive effects of ketamine are relatively unknown. This study aims to examine the neurocognitive effects of six ketamine infusions and the association of baseline neurocognitive function and the change in severity of depressive symptoms after the last infusions. METHODS Sixty-four patients with MDD completed six intravenous infusions of ketamine (0.5 mg/kg over 40 min) administered over a 12-day period (Monday-Wednesday-Friday), and were followed by a 2-week observational period. Four domains of neurocognitive function (including speed of processing, working memory, visual learning and verbal learning) were assessed using the MATRICS Consensus Cognitive Battery (MCCB) at 0, 13 and 26 days. RESULTS In linear mixed model, significant improvements were found in terms of speed of processing (F = 20.7, p < 0.001) and verbal learning (F = 11.1, p < 0.001). The Sobel test showed the improvement of speed of processing (Sobel test = 2.8, p < 0.001) and verbal learning (Sobel test = 3.6, p < 0.001) were significantly mediated by change in depressive symptoms. Other two neurocognitive domains showed no significant changes over time. Correlation analysis showed no significant association of change in depressive symptoms with neurocognitive function at baseline. CONCLUSION Our findings suggest that six ketamine infusions were associated with the improvement of speed of processing and verbal learning, which were partly accounted for by improvement in the severity of depression symptoms over time.
Collapse
Affiliation(s)
- Wei Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Yan-Ling Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Wei-Jian Liu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Cheng-Yu Wang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Yan-Ni Zhan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Han-Qiu Li
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Li-Jian Chen
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Ming-D Li
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu-Ping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China.
| |
Collapse
|
30
|
Ahrens S, Ream MA, Slaughter LA. Status Epilepticus in the Neonate: Updates in Treatment Strategies. Curr Treat Options Neurol 2019; 21:8. [PMID: 30773607 DOI: 10.1007/s11940-019-0546-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to report recent advances in treatment of neonatal seizures, with a specific focus on new literature since a 2013 systematic review performed by this author (Slaughter) and others. There is a paucity of data with regard to well-defined status epilepticus (SE) in neonates, so treatment of recurrent seizures was also included in this inquiry. We aimed to summarize the efficacy and safety profiles of current therapeutic options as well as describe trends in medication selection in the neonatal intensive care unit (NICU) setting. RECENT FINDINGS Phenobarbital remains first-line therapy in practice, though there is increasing evidence of its neurotoxicity and long-term sequelae. Bumetanide failed an open-label trial for efficacy, demonstrated an increased risk for hearing loss, and has since fallen out of favor for use in this population. New agents, such as levetiracetam and topiramate, still have very limited data but appear to be as efficacious as older medications, with more favorable side effect profiles. There are limited high-level evidence-based data to guide treatment of neonatal seizures. Emerging research focusing on drug mechanisms and safety profiles may provide additional information to guide decisions; however, further research is needed.
Collapse
Affiliation(s)
- Stephanie Ahrens
- Division of Neurology, Department of Pediatrics, Nationwide Children's Hospital, Ohio State University, 611 E Livingston Avenue FB4, Columbus, OH, 43205, USA.
| | - Margie A Ream
- Division of Neurology, Department of Pediatrics, Nationwide Children's Hospital, Ohio State University, 611 E Livingston Avenue FB4, Columbus, OH, 43205, USA
| | - Laurel A Slaughter
- Division of Neurology, Department of Pediatrics, Nationwide Children's Hospital, Ohio State University, 611 E Livingston Avenue FB4, Columbus, OH, 43205, USA
| |
Collapse
|
31
|
Paediatric Pain Medicine: Pain Differences, Recognition and Coping Acute Procedural Pain in Paediatric Emergency Room. ACTA ACUST UNITED AC 2018; 54:medicina54060094. [PMID: 30486427 PMCID: PMC6306713 DOI: 10.3390/medicina54060094] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/01/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022]
Abstract
Paediatric pain and its assessment and management are challenging for medical professionals, especially in an urgent care environment. Patients in a paediatric emergency room (PER) often undergo painful procedures which are an additional source of distress, anxiety, and pain. Paediatric procedural pain is often underestimated and neglected because of various myths, beliefs, and difficulties in its evaluation and treatment. However, it is very different from other origins of pain as it can be preventable. It is known that neonates and children can feel pain and that it has long-term effects that last through childhood into adulthood. There are a variety of pain assessment tools for children and they should be chosen according to the patient’s age, developmental stage, communication skills, and medical condition. Psychological factors such as PER environment, preprocedural preparation, and parental involvement should also be considered. There are proven methods to reduce a patient’s pain and anxiety during different procedures in PER. Distraction techniques such as music, videogames, virtual reality, or simple talk about movies, friends, or hobbies as well as cutaneous stimulation, vibration, cooling sprays, or devices are effective to alleviate procedural pain and anxiety. A choice of distraction technique should be individualized, selecting children who could benefit from nonpharmacological pain treatment methods or tools. Nonpharmacological pain management may reduce dosage of pain medication or exclude pharmacological pain management. Most nonpharmacological treatment methods are cheap, easily accessible, and safe to use on every child, so it should always be a first choice when planning a patient’s care. The aim of this review is to provide a summary of paediatric pain features, along with their physiology, assessment, management, and to highlight the importance and efficacy of nonpharmacological pain management in an urgent paediatric care setting.
Collapse
|
32
|
Golub D, Yanai A, Darzi K, Papadopoulos J, Kaufman B. Potential consequences of high-dose infusion of ketamine for refractory status epilepticus: case reports and systematic literature review. Anaesth Intensive Care 2018; 46:516-528. [PMID: 30189827 DOI: 10.1177/0310057x1804600514] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our goal was to provide comprehensive data on the effectiveness of ketamine in refractory status epilepticus (RSE) and to describe the potential consequences of long-term ketamine infusion. Ketamine, an N-methyl D-aspartate (NMDA) receptor antagonist, blocks excitatory pathways contributing to ongoing seizure. While ketamine use is standard in anaesthetic induction, no definitive protocol exists for its use in RSE, and little is known about its adverse effects in long-term, high-dose administration. We present two cases of RSE that responded rapidly to ketamine infusion, both with fatal outcomes secondary to metabolic acidosis and cardiovascular collapse. We performed a systematic review of the application and consequences of ketamine use in RSE. PubMed, Ovid, MEDLINE and PMC were searched for articles describing ketamine treatment for RSE according to a predetermined search strategy and inclusion criteria. The systematic review revealed wide discrepancies in ketamine dosing (infusion maintenance dose range 0.0075-10.5 mg/kg/hour), but good outcomes in medically managed RSE (75% of studies reported moderate or complete seizure control in adults, 62.5% in paediatrics). Additionally, literature review elucidated a potentially causal relationship between prolonged ketamine infusion and both cardiovascular and metabolic dysregulation. Ketamine is effective in RSE by antagonising excitotoxic NMDA receptors. However, there is high variability in ketamine dosing and scarce data on its safety in long-term infusion. Metabolic acidosis and haemodynamic instability associated with the use of long-term, high-dose ketamine infusions must be of concern to clinicians administering ketamine to critically ill patients.
Collapse
Affiliation(s)
| | | | | | | | - B Kaufman
- Professor, Departments of Medicine, Anesthesiology, Neurology and Neurosurgery, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
33
|
Olsson E, Anderzén-Carlsson A, Atladóttir SM, Axelin A, Campbell-Yeo M, Eriksson M, Kristjánsdóttir G, Peltonen E, Stevens B, Vederhus B, Andersen RD. Cultural adaptation and harmonization of four Nordic translations of the revised Premature Infant Pain Profile (PIPP-R). BMC Pediatr 2018; 18:349. [PMID: 30409118 PMCID: PMC6225673 DOI: 10.1186/s12887-018-1322-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 10/24/2018] [Indexed: 02/06/2023] Open
Abstract
Background Preterm infants are especially vulnerable to pain. The intensive treatment often necessary for their survival unfortunately includes many painful interventions and procedures. Untreated pain can lead to both short- and long-term negative effects. The challenge of accurately detecting pain has been cited as a major reason for lack of pain management in these non-verbal patients. The Premature Infant Pain Profile (PIPP) is one of the most extensively validated measures for assessing procedural pain in premature infants. A revised version, PIPP-R, was recently published and is reported to be more user-friendly and precise than the original version. The aims of the study were to develop translated versions of the PIPP-R in Finnish, Icelandic, Norwegian, and Swedish languages, and to establish their content validity through a cultural adaptation process using cognitive interviews. Methods PIPP-R was translated using the recommendations from the International Society for Pharmacoeconomics and Outcomes Research and enhanced with cognitive interviews. The respondent nurse was given a copy of the translated, national version of the measure and used this together with a text describing the infant in the film to assess the pain of an infant in a short film. During the assessment the nurse was asked to verbalize her thought process (thinking aloud) and upon completion the interviewer administered probing questions (verbal probing) from a structured interview guide. The interviews were recorded, transcribed, and analyzed using a structured matrix approach. Results The systematic approach resulted in translated and culturally adapted versions of PIPP-R in the Finnish, Icelandic, Norwegian and Swedish languages. During the cultural adaptation process several problems were discovered regarding how the respondent understood and utilized the measure. The problems were either measure problems or other problems. Measure problems were solved by a change in the translated versions of the measure, while for other problems different solutions such as education or training were suggested. Conclusions This study have resulted in translations of the PIPP-R that have content validity, high degree of clinical utility and displayed beginning equivalence with each other and the original version of the measure.
Collapse
Affiliation(s)
- Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University Hospital, S-701 85, Örebro, Sweden. .,Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden.
| | - Agneta Anderzén-Carlsson
- University Health Care Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Sigríður María Atladóttir
- Faculty of Nursing, University of Iceland, Reykjavik, Iceland.,Neonatal Intensive Care Unit, Lanspitali University Children's Hospital, Reykjavik, Iceland
| | - Anna Axelin
- Department of Nursing Science, University of Turku, Turku, Finland
| | - Marsha Campbell-Yeo
- School of Nursing, Faculty of Health Professions and Departments of Pediatrics, Psychology & Neuroscience, Dalhousie University, Halifax, Canada.,Centre for Pediatric Pain Research, IWK Health Centre, Halifax, Canada
| | - Mats Eriksson
- Faculty of Medicine and Health, School of Health Sciences, Örebro University, Örebro, Sweden
| | - Guðrún Kristjánsdóttir
- Faculty of Nursing, University of Iceland, Reykjavik, Iceland.,Neonatal Intensive Care Unit, Lanspitali University Children's Hospital, Reykjavik, Iceland
| | - Emilia Peltonen
- Department of Nursing Science, University of Turku, Turku, Finland
| | - Bonnie Stevens
- Lawrence S Bloomberg, Faculty of Nursing, University of Toronto, Toronto, Canada.,Department of Nursing, The Hospital for Sick Children, Toronto, Canada
| | - Bente Vederhus
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Randi Dovland Andersen
- Department of Child and Adolescent Health Services, Telemark Hospital, Skien, Norway.,Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Heiberger AL, Ngorsuraches S, Olgun G, Luze L, Leimbach C, Madison H, Lakhani SA. Safety and Utility of Continuous Ketamine Infusion for Sedation in Mechanically Ventilated Pediatric Patients. J Pediatr Pharmacol Ther 2018; 23:447-454. [PMID: 30697129 DOI: 10.5863/1551-6776-23.6.447] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVES The selection of sedative medications for mechanically ventilated pediatric patients remains an ongoing clinical challenge. Although continuous ketamine infusion has been used in this population, support for its use remains largely anecdotal. This study describes a single institution's use of ketamine infusions as part of a sedation protocol in the pediatric intensive care unit (PICU). METHODS This was a retrospective study of children who received ketamine infusions as part of a multidrug sedation protocol in a 12-bed PICU at a tertiary children's hospital. Outcomes included effectiveness of ketamine infusion in providing adequate sedation as determined by State Behavioral Scale (SBS) scores and incidence of adverse events. RESULTS A total of 22 children receiving ketamine continuous infusion as part of a multidrug sedation protocol from February 2014 through October 2015 were eligible and enrolled in the study. Ketamine continuous infusion was administered in addition to at least 2 other sedation infusions at an average rate of 1.02 ± 0.50 mg/kg/hr, with a range of 0.07 to 2.0 mg/kg/hr. The duration of ketamine was 65.7 ± 41.01 hours, with a range of 19 to 153 hours. There was no significant change in SBS scores before and after initiation of ketamine infusion. Although not statistically significant, patients with inadequate sedation prior to starting ketamine required fewer bolus sedation doses and had improved sedation after ketamine was started. There were no reported adverse events. CONCLUSIONS The addition of a ketamine infusion as part of a multidrug sedation regimen was at least as effective as patients' regimen prior to ketamine addition in this population of intubated pediatric patients, with no adverse events.
Collapse
|
35
|
Liu FF, Zhao S, Liu P, Huo SP. Influence of mTOR signaling pathway on ketamine-induced injuries in the hippocampal neurons of rats. Neurol Res 2018; 41:77-86. [PMID: 30373500 DOI: 10.1080/01616412.2018.1531203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To explore the influences of mammalian target of rapamycin (mTOR) signaling pathway on ketamine-induced apoptosis, oxidative stress and Ca2+ concentration in the hippocampal neurons of rats. METHODS The primary hippocampal neurons isolated from fetal Sprague Dawley rats were treated with ketamine (0, 50, 100 and 500 μM) for 4 days to observe its effect on mTOR signaling pathway and apoptosis of rat hippocampal neurons. Then, the hippocampal neurons were divided into C (Control), R (Rapamycin, an inhibitor of mTOR signaling pathway), K (Ketamine) and R + K (Rapamycin + Ketamine) groups to detect the apoptosis, reactive oxygen species (ROS) production, and Ca2+ concentration via the terminal transferase uridyl nick end labelling (TUNEL) assay, dichloro-dihydro-fluorescein diacetate (DCFH-DA) method and Fluo-3 acetoxymethyl ester (Fluo-3AM) staining, respectively. The expressions of mTOR signaling pathway and apoptosis-related proteins in hippocampal neurons were examined by qRT-PCR and Western blot. RESULTS Ketamine could dose-dependently promote the apoptosis of rat hippocampal neurons with upregulation of p-mTOR and its downstream regulators (p-4E-BP-1 and p-p70S6K). However, ketamine-induced apoptosis in hippocampal neurons was reversed significantly by the administration of rapamycin, as evident by the decrease in expressions of pro-apoptotic proteins (Bax and cleaved Caspase-3) and the increase in anti-apoptotic protein (Bcl-2). Meanwhile, the ROS generation and Ca2+ concentration was inhibited accompanied with reduced malonildialdehyde levels but elevated superoxide and glutathione peroxidase activities. CONCLUSION Inhibition of mTOR signaling pathway protected rat hippocampal neurons from ketamine-induced injuries via reducing apoptosis, oxidative stress, as well as Ca2+ concentration. ABBREVIATIONS mTOR: mammalian target of rapamycin; SD: Sprague-Dawley; SPF: Specific-pathogen free; ROS: reactive oxygen species; TUNEL: terminal transferase uridyl nick end labelling; DCFH-DA: Dichloro-dihydro-fluorescein diacetate; Fluo-3A: Fluo-3 acetoxymethyl ester; NMDAR: non-competitive N-methyl-D-aspartame glutamate receptor; 4E-BP1: 4E binding protein 1; p70S6K: p70 S6 Kinase; PCR: Polymerase chain reaction; MDA: malonildialdehyde; GSH-PX: glutathione peroxidase; ANOVA: One-way Analysis of Variance.
Collapse
Affiliation(s)
- Fei-Fei Liu
- a Department of Anesthesiology , Third Hospital of Hebei Medical University , Shijiazhuang , China
| | - Shuang Zhao
- a Department of Anesthesiology , Third Hospital of Hebei Medical University , Shijiazhuang , China
| | - Peng Liu
- a Department of Anesthesiology , Third Hospital of Hebei Medical University , Shijiazhuang , China
| | - Shu-Ping Huo
- a Department of Anesthesiology , Third Hospital of Hebei Medical University , Shijiazhuang , China
| |
Collapse
|
36
|
Jiang JD, Zheng XC, Huang FY, Gao F, You MZ, Zheng T. MicroRNA-107 regulates anesthesia-induced neural injury in embryonic stem cell derived neurons. IUBMB Life 2018; 71:20-27. [PMID: 30308117 PMCID: PMC7379593 DOI: 10.1002/iub.1911] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 05/07/2018] [Indexed: 01/11/2023]
Abstract
Ketamine, though widely used in pediatric anesthesia, may induce cortical neurotoxicity in young patients. This study focused on an in vitro model of rat brain embryonic stem cell (ESC)-derived neurons to investigate the effects of microRNA-107 (miR-107) on ketamine-induced neural injury. Rat brain ESCs were proliferated in vitro and differentiated toward neuronal fate. Ketamine induced neural injury in ESC-derived neurons was inspected by TUNEL and neurite growth assays. Ketamine-induce aberrant miR-107 expression was examined by qRT-PCR. MiR-107 was downregulated in ESCs through lentiviral transduction. Its effect on ketamine-induced neural injury in ESC-derived neurons was then examined. Potential downstream target of miR-107, brain derived neurotrophin factor (BDNF), was inspected by dual-luciferase reporter assay and qRT-PCR. BDNF was knocked down, through siRNA transfection, in NSCs to investigate its functional involvement in miR-107 mediated neural protection in ketamine-injured NSC-derived neurons. Ketamine induced apoptosis, neurite degeneration, and upregulated miR-107 in NSC-derived neurons. Lentivirus-mediated miR-107 downregulation attenuated ketamine-induced neural injury. BDNF was proven to be directly and inversely regulated by miR-107 in NSC-derived neurons. SiRNA-mediated BDNF inhibition reversed the protective effect of miR-107 downregulation on ketamine injury in NSC-derived neurons. MiR-107 / BDNF was demonstrated to be an important epigenetic signaling pathway in regulating ketamine-induced neural injury in cortical neurons. © 2018 The Authors. IUBMB Life published by Wiley Periodicals,Inc. on behalf of International Union of Biochemistry and Molecular Biology., 71(1):20-27, 2019.
Collapse
Affiliation(s)
- Jun Dan Jiang
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Xiao Chun Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China.,Department of Anesthesiology, Fujian Provincial Emergency Center, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Feng Yi Huang
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Fei Gao
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Mei Zhen You
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| | - Ting Zheng
- Department of Anesthesiology, Fujian Provincial Hospital, Provincial Clinical Medical College, Fujian Medical University, Fujian, China
| |
Collapse
|
37
|
Shao CZ, Xia KP. Sevoflurane anesthesia represses neurogenesis of hippocampus neural stem cells via regulating microRNA-183-mediated NR4A2 in newborn rats. J Cell Physiol 2018; 234:3864-3873. [PMID: 30191980 DOI: 10.1002/jcp.27158] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/10/2018] [Indexed: 12/23/2022]
Abstract
Sevoflurane has been commonly utilized in nonobstetric surgeries in pregnant women, and its impacts on fetal brain are still not completely known. Ectopic NR4A2 expression has been reported to be related with familial Parkinson disease, and through dual luciferase we found that NR4A2 is a target gene of microRNA-183 (miR-183). We proposed a hypothesis that miR-183 may participate in the process by targeting NR4A2 in neurons after sevoflurane anesthesia. To verify the effect of sevoflurane on hippocampal neural stem cells (NSCs) proliferation and differentiation, we conducted EdU assay and immunofluorescence staining. Next, for better understanding of the impact of miR-183, we altered the miR-183 expression using mimic and inhibitor. Meanwhile, the targeting relationship between miR-183 and NR4A2 was validated by a bioinformatics website and dual-luciferase reporter gene assay. Finally, expressions of miR-184, NR4A2, SRY (sex-determining region Y)-box 2 (Sox2), and brain-derived neurotrophic factor (BDNF) were determined and evaluated by reverse transcription quantitative polymerase chain reaction and western blot analysis. First, sevoflurane was determined a crucial factor in biological behaviors of hippocampal NSCs. Moreover, upregulated miR-183 expression by mimic inhibited the proliferation and differentiation of NSCs. Sevoflurane negatively regulated NR4A2 and Sox2 expressions but positively regulated miR-183 and BDNF expressions. Our findings revealed the underlying novel mechanism by which sevoflurane inhibits hippocampal NSC proliferation and differentiation through interaction with miR-183 and NR4A2. The study provides reliable reference for safe application of sevoflurane anesthesia in neonates.
Collapse
Affiliation(s)
- Chang-Zhong Shao
- Department of Anesthesiology, Linyi People's Hospital Affiliated to Shandong University, Linyi, China
| | - Kun-Peng Xia
- Department of Anesthesiology, Linyi People's Hospital Affiliated to Shandong University, Linyi, China
| |
Collapse
|
38
|
Höfler J, Trinka E. Intravenous ketamine in status epilepticus. Epilepsia 2018; 59 Suppl 2:198-206. [DOI: 10.1111/epi.14480] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Julia Höfler
- Department of Neurology; Paracelsus Medical University Salzburg and Christian Doppler Medical Center; Salzburg Austria
| | - Eugen Trinka
- Department of Neurology; Paracelsus Medical University Salzburg and Christian Doppler Medical Center; Salzburg Austria
| |
Collapse
|
39
|
Bourgoin L, Caeymaex L, Decobert F, Jung C, Danan C, Durrmeyer X. Administering atropine and ketamine before less invasive surfactant administration resulted in low pain scores in a prospective study of premature neonates. Acta Paediatr 2018. [PMID: 29532502 DOI: 10.1111/apa.14317] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
AIM Less invasive surfactant administration (LISA) can avoid tracheal intubation for neonatal respiratory distress syndrome, but can be painful because it requires laryngoscopy. The aim of this study was to assess the efficacy and tolerance of intravenous atropine plus ketamine administration before LISA. METHODS We conducted a prospective observational study of all premature infants hospitalised in our French neonatal intensive care unit treated with LISA between March 2015 and March 2016. Ketamine was titrated by 0.5 mg/kg increments. The technical conditions, pain scores, emergent intubations and vital signs were collected and analysed. RESULTS Values are reported as medians (interquartile ranges). We included 29 patients with a gestational age of 29.6 (28.6-30.9) weeks and birth weight of 1290 (945-1600) grams. Technical conditions were satisfying for 24 infants (83%). The Faceless Acute Neonatal Pain Scale score was 2 (2-4); seven infants (24%) required tracheal intubation before LISA could be performed; 17 (59%) had a pulse oxymetry value under 80% that lasted more than 60 seconds. Heart rate and mean arterial blood pressure transiently increased. CONCLUSION Atropine plus ketamine before LISA resulted in low pain scores and stable haemodynamic parameters, but prolonged desaturations or apnoea leading to tracheal intubation were frequently observed.
Collapse
Affiliation(s)
- L Bourgoin
- Neonatal Intensive Care Unit; Centre Hospitalier Intercommunal de Créteil; Créteil France
| | - L Caeymaex
- Neonatal Intensive Care Unit; Centre Hospitalier Intercommunal de Créteil; Créteil France
| | - F Decobert
- Neonatal Intensive Care Unit; Centre Hospitalier Intercommunal de Créteil; Créteil France
- Inserm U955, Team 13; Université Paris Est (UPEC); Créteil France
| | - C Jung
- Clinical Research Center; Centre Hospitalier Intercommunal de Créteil; Créteil France
| | - C Danan
- Neonatal Intensive Care Unit; Centre Hospitalier Intercommunal de Créteil; Créteil France
- Inserm U955, Team 13; Université Paris Est (UPEC); Créteil France
- Clinical Research Center; Centre Hospitalier Intercommunal de Créteil; Créteil France
| | - X Durrmeyer
- Neonatal Intensive Care Unit; Centre Hospitalier Intercommunal de Créteil; Créteil France
- Inserm U955, Team 13; Université Paris Est (UPEC); Créteil France
| |
Collapse
|
40
|
Liao PH, Yang WK, Yang CH, Lin CH, Hwang CC, Chen PJ. Illicit drug ketamine induces adverse effects from behavioral alterations and oxidative stress to p53-regulated apoptosis in medaka fish under environmentally relevant exposures. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 237:1062-1071. [PMID: 29146197 DOI: 10.1016/j.envpol.2017.11.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/27/2017] [Accepted: 11/06/2017] [Indexed: 06/07/2023]
Abstract
With increasing problems of drug abuse worldwide, aquatic ecosystems are contaminated by human pharmaceuticals from the discharge of hospital or municipal effluent. However, ecotoxicity data and related toxic mechanism for neuroactive controlled or illicit drugs are still lacking, so assessing the associated hazardous risk is difficult. This study aims to investigate the behavioral changes, oxidative stress, gene expression and neurotoxic or apoptosis effect(s) in larvae of medaka fish (Oryzias latipes) with environmentally relevant exposures of ketamine (KET) solutions for 1-14 days. KET exposure at an environmentally relevant concentration (0.004 μM) to 40 μM conferred specific patterns in larval swimming behavior during 24 h. At 14 days, such exposure induced dose- and/or time-dependent alteration on reactive oxygen species induction, the activity of antioxidants catalase and superoxide dismutase, glutathione S-transferase and malondialdehyde contents in fish bodies. KET-induced oxidative stress disrupted the expression of acetylcholinesterase and p53-regulated apoptosis pathways and increased caspase expression in medaka larvae. The toxic responses of medaka larvae, in terms of chemical effects, were qualitatively analogous to those of zebrafish and mammals. Our results implicate a toxicological impact of waterborne KET on fish development and human health, for potential ecological risks of directly releasing neuroactive drugs-containing wastewater into the aquatic environment.
Collapse
Affiliation(s)
- Pei-Han Liao
- Department of Agricultural Chemistry, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Wen-Kai Yang
- Department of Agricultural Chemistry, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsin Yang
- Department of Agricultural Chemistry, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Chun-Hon Lin
- Department of Agricultural Chemistry, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Chin-Chu Hwang
- Department of Agricultural Chemistry, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Pei-Jen Chen
- Department of Agricultural Chemistry, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
41
|
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
|
42
|
NMDA-receptor Antagonism in Pediatric Pancreatitis: Use of Ketamine and Methadone in a Teenager With Refractory Pain. J Pediatr Gastroenterol Nutr 2018; 66:e134-e136. [PMID: 29394214 DOI: 10.1097/mpg.0000000000001907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
43
|
Prospective follow-up of a cohort of preterm infants<33 WG receiving ketamine for tracheal intubation in the delivery room: Neurological outcome at 1 and 2 years. Arch Pediatr 2018; 25:295-300. [PMID: 29628409 DOI: 10.1016/j.arcped.2018.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/23/2018] [Accepted: 03/04/2018] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Although ketamine analgesia is effective in reducing pain and facilitating the tracheal intubation of newborns in the delivery room, no data on the neurological effects of this treatment are available. This study compared the neurodevelopmental outcomes at 2 years of age in a cohort of preterm newborns having received ketamine prior to tracheal intubation at birth (the ketamine group) and in a control group. METHODS We included newborns delivered at less than 33 weeks gestational age (WGA) having undergone tracheal intubation at birth. The Ages and Stages Questionnaire (ASQ) was completed at 1 and 2 years of age. The development quotient (DQ) was calculated from the revised Brunet-Lezine score assessed at a corrected age of 2 years. RESULTS There were no statistically significant differences between the ketamine group (n=54 at 1 year and n=51 at 2 years) and the control group (n=16 at 1 and 2 years) in terms of the mean±standard deviation DQ at the age of 2 (98±12 vs. 103±9, respectively; P=0.17) and the ASQ score at the age of 2 (221±44 vs. 230±39, respectively; P=0.55). DISCUSSION This prospective cohort of 51 preterm newborns having received ketamine at birth did not reveal any differences in terms of neurological development at the age of 2 (relative to a control group and the literature data). These preliminary results must be confirmed in a randomized trial with longer follow-up.
Collapse
|
44
|
Jiang S, Li X, Jin W, Duan X, Bo L, Wu J, Zhang R, Wang Y, Kang R, Huang L. Ketamine-induced neurotoxicity blocked by N-Methyl-d-aspartate is mediated through activation of PKC/ERK pathway in developing hippocampal neurons. Neurosci Lett 2018; 673:122-131. [PMID: 29501685 DOI: 10.1016/j.neulet.2018.02.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/12/2018] [Accepted: 02/23/2018] [Indexed: 11/26/2022]
Abstract
Ketamine, a non-competitive N-methyl d-aspartate (NMDA) receptor antagonist, is widely used in pediatric clinical practice. However, prolonged exposure to ketamine results in widespread anesthetic neurotoxicity and long-term neurocognitive deficits. The molecular mechanisms that underlie this important event are poorly understood. We investigated effects of anesthetic ketamine on neuroapoptosis and further explored role of NMDA receptors in ketamine-induced neurotoxicity. Here we demonstrate that ketamine induces activation of cell cycle entry, resulting in cycle-related neuronal apoptosis. On the other hand, ketamine administration alters early and late apoptosis of cultured hippocampus neurons by inhibiting PKC/ERK pathway, whereas excitatory NMDA receptor activation reverses these effects. Ketamine-induced neurotoxicity blocked by NMDA is mediated through activation of PKC/ERK pathway in developing hippocampal neurons.
Collapse
Affiliation(s)
- Sufang Jiang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xuze Li
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Wei Jin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei Province, China
| | - Xiaofeng Duan
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Lijun Bo
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jiangli Wu
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Rui Zhang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ying Wang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Rongtian Kang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Lining Huang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
45
|
Shi Y, Li J, Chen C, Xia Y, Li Y, Zhang P, Xu Y, Li T, Zhou W, Song W. Ketamine Modulates Zic5 Expression via the Notch Signaling Pathway in Neural Crest Induction. Front Mol Neurosci 2018; 11:9. [PMID: 29472839 PMCID: PMC5810301 DOI: 10.3389/fnmol.2018.00009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/08/2018] [Indexed: 12/23/2022] Open
Abstract
Ketamine is a potent dissociative anesthetic and the most commonly used illicit drug. Many addicts are women at childbearing age. Although ketamine has been extensively studied as a clinical anesthetic, its effects on embryonic development are poorly understood. Here, we applied the Xenopus model to study the effects of ketamine on development. We found that exposure to ketamine from pre-gastrulation (stage 7) to early neural plate (stage 13.5) resulted in disruption of neural crest (NC) derivatives. Ketamine exposure did not affect mesoderm development as indicated by the normal expression of Chordin, Xbra, Wnt8, and Fgf8. However, ketamine treatment significantly inhibited Zic5 and Slug expression at early neural plate stage. Overexpression of Zic5 rescued ketamine-induced Slug inhibition, suggesting the blockage of NC induction was mediated by Zic5. Furthermore, we found Notch signaling was altered by ketamine. Ketamine inhibited the expression of Notch targeted genes including Hes5.2a, Hes5.2b, and ESR1 and ketamine-treated embryos exhibited Notch-deficient somite phenotypes. A 15 bp core binding element upstream of Zic5 was induced by Notch signaling and caused transcriptional activation. These results demonstrated that Zic5 works as a downstream target gene of Notch signaling in Xenopus NC induction. Our study provides a novel teratogenic mechanism whereby ketamine disrupts NC induction via targeting a Notch-Zic5 signaling pathway.
Collapse
Affiliation(s)
- Yu Shi
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jiejing Li
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Chunjiang Chen
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yongwu Xia
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanxi Li
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Pan Zhang
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Xu
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tingyu Li
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihui Zhou
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihong Song
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
46
|
Lin28b and Sox2 regulate anesthesia-induced neural degeneration in neural stem cell derived neurons. Eur J Pharmacol 2018; 820:167-172. [DOI: 10.1016/j.ejphar.2017.12.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/10/2017] [Accepted: 12/11/2017] [Indexed: 01/22/2023]
|
47
|
Effects on adult cognitive function after neonatal exposure to clinically relevant doses of ionising radiation and ketamine in mice. Br J Anaesth 2018; 120:546-554. [PMID: 29452811 DOI: 10.1016/j.bja.2017.11.099] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 10/31/2017] [Accepted: 11/10/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Radiological methods for screening, diagnostics and therapy are frequently used in healthcare. In infants and children, anaesthesia/sedation is often used in these situations to relieve the patients' perception of stress or pain. Both ionising radiation (IR) and ketamine have been shown to induce developmental neurotoxic effects and this study aimed to identify the combined effects of these in a murine model. METHODS Male mice were exposed to a single dose of ketamine (7.5 mg kg-1 body weight) s.c. on postnatal day 10. One hour after ketamine exposure, mice were whole body irradiated with 50-200 mGy gamma radiation (137Cs). Behavioural observations were performed at 2, 4 and 5 months of age. At 6 months of age, cerebral cortex and hippocampus tissue were analysed for neuroprotein levels. RESULTS Animals co-exposed to IR and ketamine displayed significant (P≤0.01) lack of habituation in the spontaneous behaviour test, when compared with controls and single agent exposed mice. In the Morris Water Maze test, co-exposed animals showed significant (P≤0.05) impaired learning and memory capacity in both the spatial acquisition task and the relearning test compared with controls and single agent exposed mice. Furthermore, in co-exposed mice a significantly (P≤0.05) elevated level of tau protein in cerebral cortex was observed. Single agent exposure did not cause any significant effects on the investigated endpoints. CONCLUSION Co-exposure to IR and ketamine can aggravate developmental neurotoxic effects at doses where the single agent exposure does not impact on the measured variables. These findings show that estimation of risk after paediatric low-dose IR exposure, based upon radiation dose alone, may underestimate the consequences for this vulnerable population.
Collapse
|
48
|
Zhang J, Cui C, Li Y, Xu H. Inhibition of GSK-3beta Signaling Pathway Rescues Ketamine-Induced Neurotoxicity in Neural Stem Cell-Derived Neurons. Neuromolecular Med 2017; 20:54-62. [PMID: 29218434 DOI: 10.1007/s12017-017-8472-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/30/2017] [Indexed: 01/13/2023]
Abstract
Clinical application of anesthetic reagent, ketamine (Keta), may induce irreversible neurotoxicity in central nervous system. In this work, we utilized an in vitro model of neural stem cells-derived neurons (nSCNs) to evaluate the role of GSK-3 signaling pathway in Keta-induced neurotoxicity. Embryonic mouse-brain neural stem cells were differentiated into neurons in vitro. Keta (50 μM)-induced neurotoxicity in cultured nSCNs was monitored by apoptosis, immunohistochemical and western blot assays, respectively. GSK-3 signaling pathways, including GSK-3α and GSK-3β, were inhibited by siRNA in the culture. The subsequent effects of GSK-3α or GSK-3β downregulation on Keta-induced neurotoxicity, including apoptosis and neurite loss, were then evaluated in nSCNs. Finally, caspase and Akt/ERK signal pathways were further examined by western blot to evaluate the regulatory effect of GSK-3 signaling pathways on Keta-induced neural injury. Keta (50 μM) caused markedly nSCN apoptosis and neurite degeneration in vitro. Keta decreased GSK-3β phosphorylation, but had no effect on GSK-3α phosphorylation. SiRNA-induced GSK-3β downregulation rescued Keta-induced neurotoxicity in nSCNs by reducing neuronal apoptosis and preventing neurite degeneration. On the other hand, GSK-3α downregulation had no effect on Keta-induced neurotoxicity. Western blot showed that, in Keta-injured nSCNs, GSK-3β downregulation reduced Caspase-1/3 proteins, but left phosphorylated Akt/ERK unchanged. GSK-3β, not GSK-3α, was specifically involved in the process of Keta-induced neurotoxicity in nSCNs. Inhibiting GSK-3β may be an effective approach to counter toxic effect of ketamine on central neurons in clinical and experimental applications.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Changlei Cui
- Department of Anesthesia, The First Hospital of Jilin University, 71 Xinming St., Chaoyang District, Changchun, 130021, China
| | - Yanhui Li
- Department of Anesthesia, The First Hospital of Jilin University, 71 Xinming St., Chaoyang District, Changchun, 130021, China
| | - Haiyang Xu
- Department of Anesthesia, The First Hospital of Jilin University, 71 Xinming St., Chaoyang District, Changchun, 130021, China.
| |
Collapse
|
49
|
Piersigilli F, Di Pede A, Catena G, Lozzi S, Auriti C, Bersani I, Capolupo I, Lipreri A, Di Ciommo V, Dotta A, Sgrò S. Propofol and fentanyl sedation for laser treatment of retinopathy of prematurity to avoid intubation. J Matern Fetal Neonatal Med 2017; 32:517-521. [DOI: 10.1080/14767058.2017.1383379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Fiammetta Piersigilli
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Alessandra Di Pede
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Gino Catena
- Department of Ophthalmology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Simona Lozzi
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Cinzia Auriti
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Iliana Bersani
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Irma Capolupo
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Anna Lipreri
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Vincenzo Di Ciommo
- Unit of Clinical Epidemiology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Andrea Dotta
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Stefania Sgrò
- Department of Anesthesiology, Bambino Gesù Children’s Hospital, Rome, Italy
| |
Collapse
|
50
|
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
|