1
|
Mashaw SA, Anwar AI, Vu JN, Thomassen AS, Beesley ML, Shekoohi S, Kaye AD. Novel and Emerging Treatments for Agitation in Schizophrenia and Bipolar Disorder. Healthcare (Basel) 2025; 13:932. [PMID: 40281882 PMCID: PMC12027268 DOI: 10.3390/healthcare13080932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Agitation is a frequent and challenging symptom in schizophrenia and bipolar disorder, characterized by heightened motor activity, emotional distress, and potential aggression. This symptom is most observed during acute episodes, representing a significant burden on patients, caregivers, and healthcare systems. Agitation is a leading cause of emergency department visits and psychiatric hospitalizations, necessitating prompt and effective interventions to ensure safety and mitigate its far-reaching impact. Traditional treatments, including high-potency antipsychotics and benzodiazepines, remain first-line options but are associated with significant drawbacks such as sedation, extrapyramidal symptoms, tolerance, and limited applicability in certain patient populations, especially those with respiratory or cardiac depression and the elderly. Non-pharmacologic strategies like de-escalation techniques and environmental modifications are invaluable but may be impractical in acute care settings, as speed and efficiency are critical in emergent settings. These limitations, including the onset of extrapyramidal symptoms with high-dose antipsychotics and the development of tolerance with benzodiazepines, highlight gaps in care, including the need for faster-acting, safer, and more patient-friendly alternatives that reduce reliance on physical restraints and invasive interventions. Methods: This review explores the evolution of treatments for agitation, focusing on alternative and innovative approaches. To highlight these treatments, an extensive review of the literature was conducted utilizing PubMed, Google Scholar, Embase.com, and other search engines. Results: Key developments include sublingual dexmedetomidine, recently FDA-approved, which offers sedation without respiratory depression and a non-invasive administration route. Similarly, subcutaneous olanzapine provides a more convenient alternative to intramuscular injections, reducing injection-related complications. Other emerging treatments such as gabapentin, pregabalin, and ketamine show promise in addressing agitation in specific contexts, including comorbid conditions and treatment-resistant cases. A comparative analysis of these therapies highlights their mechanisms of action, clinical evidence, and practical challenges. Conclusions: Future directions emphasize intranasal delivery systems, novel pharmacologic agents, and potential roles for cannabinoids in managing agitation. These innovations aim to balance rapid symptom control with improved patient safety and experience. The set back with these emerging techniques is a lack of standardized dosing and protocols. They also face ethical concerns, including the chance of misuse or abuse, as well as regulatory barriers, as they lack FDA approval and their legality changes between states. This review underscores the clinical, practical, and ethical considerations in advancing care for agitated patients, paving the way for more effective and compassionate management strategies in psychiatric settings.
Collapse
Affiliation(s)
- Sydney A. Mashaw
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA 71103, USA
| | - Ahmed I. Anwar
- Department of Psychology, Quinnipiac University, Hamden, CT 06518, USA
| | - Judy N. Vu
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA 71103, USA
| | - Austin S. Thomassen
- School of Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA 71103, USA
| | - Maya L. Beesley
- College of Natural Sciences and Mathematics, University of Denver, 2199 S University Blvd, Denver, CO 80210, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA 71103, USA
| | - Alan D. Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA 71103, USA
- Departments of Pharmacology, Toxicology, and Neurosciences, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA 71103, USA
| |
Collapse
|
2
|
Yan Y, Zhang Y, Liu M, Li L, Zheng Y. Neuroprotection vs. Neurotoxicity: The Dual Impact of Brain Lipids in Depression. Int J Mol Sci 2025; 26:2722. [PMID: 40141364 PMCID: PMC11943007 DOI: 10.3390/ijms26062722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/03/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Growing neurochemical evidence highlights cerebral lipid dysregulation as a key factor in the pathophysiology of major depressive disorder (MDD). This review systematically explores the dual roles of lipid species in both normal behavioral regulation and MDD development. By critically examining the recent literature, we classify these lipid species into two functional categories based on their functional neuroactivity: (1) neuroprotective lipids (sphingomyelin, cholesterol, cardiolipin, sphingosine, phosphatidic acid, and phosphatidylserine), which exert neuroprotective effects by modulating membrane fluidity and supporting synaptic vesicle trafficking; and (2) neurotoxic lipids (ceramides, phosphatidylinositol, phosphocholine, and phosphatidylethanolamine), which promote apoptotic signaling cascades and disrupt mitochondrial bioenergetics. An unresolved but critical question pertains to the maintenance of homeostatic equilibrium between these opposing lipid classes. This balance is essential, given their significant impact on membrane protein localization and function, monoaminergic neurotransmitter metabolism, energy homeostasis, and redox balance in neural circuits involved in mood regulation. This emerging framework positions cerebral lipidomics as a promising avenue for identifying novel therapeutic targets and developing biomarker-based diagnostic approaches for MDD treatment.
Collapse
Affiliation(s)
| | | | | | | | - Yanrong Zheng
- Zhejiang Key Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
3
|
Roncero C, Merizalde-Torres M, Szerman N, Torrens M, Vega P, Andres-Olivera P, Javier Álvarez F. Is there a risk of esketamine misuse in clinical practice? Ther Adv Drug Saf 2025; 16:20420986241310685. [PMID: 39882342 PMCID: PMC11776012 DOI: 10.1177/20420986241310685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/12/2024] [Indexed: 01/31/2025] Open
Abstract
In 2019, intranasal esketamine gained approval as a promising therapy for those individuals grappling with treatment-resistant depression. Both clinical trials and real-world studies have underscored its efficacy in alleviating and remitting depressive symptoms, with sustained benefits observed for nearly 4.5 years. As the S-enantiomer of ketamine, esketamine's dosing guidelines and strict medical supervision stem from prior research on ketamine's use in depression and history as a recreational drug. Despite initial concerns, long-term clinical studies have not documented instances of abuse, misuse, addiction or withdrawal, and the same was found in case reports or subsamples of high-risk populations with comorbidities such as substance use disorder or alcohol use disorder. Esketamine has proven to be safe and well tolerated without fostering new-onset substance use in vulnerable groups. Real-world studies reinforced these observations, reporting no adverse events (AEs) related to pharmacological interactions of esketamine with any other substance, and no new-onset drug or alcohol misuse, craving, misuse or diversion of use. Reports of esketamine craving remain rare, with only one case report documented in 2022. Most drug-related AEs reported in pharmacovigilance databases are those identified in the product's technical data sheet and with known reported frequency. More importantly, no register of illicit acquisition of esketamine or its tampering for obtaining ketamine or other altered products was found in our search. Overall, our review confirms esketamine's safety across diverse patient populations, reassuring its responsible use and the scarcity of reports of abuse or misuse since its introduction to the market.
Collapse
Affiliation(s)
- Carlos Roncero
- Health Science Faculty, European University Miguel de Cervantes (UEMC), C/Padre Julio Chevalier, 2, 47012, Valladolid, Spain
- Psychiatry Unit, School of Medicine, University of Salamanca, Salamanca, Spain
- Network of Research in Primary Care of Addictions (RIAPAD) Instituto Carlos III, Madrid, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Milton Merizalde-Torres
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Psychiatry Service, University Healthcare Complex of Salamanca, Salamanca, Spain
| | - Néstor Szerman
- World Association on Dual Disorders. World Psychiatric Association, Section on Dual Disorders, Madrid, Spain
- Institute of Psychiatry and Mental Health, University Hospital Gregorio Marañón, Madrid, Spain
| | - Marta Torrens
- Network of Research in Primary care of Addictions (RIAPAD)
- School of Medicine, Universitat de Vic-Central de Catalunya, Vic, Spain
- Medicine and Life Sciences Department, Universitat Pompeu Fabra, Barcelona, Spain
- Addiction Research Group (GRAd), Neuroscience Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Pablo Vega
- Institute for Addictions, Madrid Salud, Madrid City Council, Madrid, Spain
| | - Pilar Andres-Olivera
- Psychiatry Unit, School of Medicine, University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Psychiatry Service, University Healthcare Complex of Salamanca, Salamanca, Spain
| | | |
Collapse
|
4
|
Wang G, Qi W, Liu QH, Guan W. GluN2A: A Promising Target for Developing Novel Antidepressants. Int J Neuropsychopharmacol 2024; 27:pyae037. [PMID: 39185814 PMCID: PMC12042802 DOI: 10.1093/ijnp/pyae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Depression is a heterogeneous disorder with high morbidity and disability rates that poses serious problems regarding mental health care. It is now well established that N-methyl D-aspartate receptor (NMDAR) modulators are being increasingly explored as potential therapeutic options for treating depression, although relatively little is known about their mechanisms of action. NMDARs are glutamate-gated ion channels that are ubiquitously expressed in the central nervous system (CNS), and they have been shown to play key roles in excitatory synaptic transmission. GluN2A, the predominant Glu2N subunit of functional NMDARs in neurons, is involved in various physiological processes in the CNS and is associated with diseases such as anxiety, depression, and schizophrenia. However, the role of GluN2A in the pathophysiology of depression has not yet been elucidated. METHODS We reviewed several past studies to better understand the function of GluN2A in depression. Additionally, we also summarized the pathogenesis of depression based on the regulation of GluN2A expression, particularly its interaction with neuroinflammation and neurogenesis, which has received considerable critical attention and is highly implicated in the onset of depression. RESULTS These evidence suggests that GluN2A overexpression impairs structural and functional synaptic plasticity, which contributes to the development of depression. Consequently, this knowledge is vital for the development of selective antagonists targeting GluN2A subunits using pharmacological and molecular methods. CONCLUSIONS Specific inhibition of the GluN2A NMDAR subunit is resistant to chronic stress-induced depressive-like behaviors, making them promising targets for the development of novel antidepressants.
Collapse
Affiliation(s)
- Gang Wang
- Department of Hepatobiliary Surgery, Zhangjiagang Hospital affiliated to Soochow University/The First People’s Hospital of Zhangjiagang City, Zhangjiagang, China
| | - Wang Qi
- Department of Pharmacology, The First People’s Hospital of Yancheng, Yancheng, China
| | - Qiu-Hua Liu
- Department of Hepatobiliary Surgery, Zhangjiagang Hospital affiliated to Soochow University/The First People’s Hospital of Zhangjiagang City, Zhangjiagang, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China
| |
Collapse
|
5
|
Nayyer MA, Khan SM, Umer M, Imran H, Khalid S, Murtaza H, Sarfraz A, Atiq N, Rasool H, Fatima M. Efficacy and safety of peri-partum Esketamine for prevention of post-partum depression in women undergoing caesarian section: A meta-analysis and systematic review of randomized controlled trials. Asian J Psychiatr 2024; 97:104090. [PMID: 38820851 DOI: 10.1016/j.ajp.2024.104090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 06/02/2024]
Abstract
Postpartum depression (PPD) is a psychiatric condition affecting women post-childbirth. Medication combined with psychotherapy, is the current protocol for its treatment. A meta-analysis was conducted using RevMan 5.4 to explore the efficacy and safety of peri-partum administration of esketamine for preventing PPD. After searching several databases to retrieve the relevant RCTs, seven were included in this analysis, with dichotomous data presented as risk ratio and continuous data as mean difference. The study found a lower incidence of PPD in the esketamine group compared to the control group (RR= 0.37), with significant difference in EPDS scores between the two groups (MD= -1.23) in the first week postpartum. The esketamine group reported a lower prevalence of PPD 4-6 weeks postpartum (RR= 0.48), and no significant difference in EPDS scores after 4 weeks postpartum (MD = -0.10). The esketamine group had a significantly higher incidence of hallucination (RR= 13.85). Other adverse effects, such as dizziness (RR= 4.09), nausea (RR= 0.88), vomiting (RR=0.74), headache (RR=1.52), nightmares (RR=1.22), pruritus (RR=0.29), and drowsiness (RR=1.57) did not show significant differences between the two groups. The study found that esketamine, with manageable side effects, reduces the prevalence of post-partum depression (PPD) after one week as well as after four to six weeks. However, the findings are limited by the limited number of available RCTs, and future research should determine the ideal dosage, the most effective method of administration and the long-term safety profile of esketamine so that it may be considered as an adjunct therapy or a potential sole treatment option.
Collapse
Affiliation(s)
| | | | | | - Haim Imran
- King Edward Medical University, Lahore, Pakistan.
| | | | | | | | - Noor Atiq
- King Edward Medical University, Lahore, Pakistan.
| | - Hamna Rasool
- King Edward Medical University, Lahore, Pakistan.
| | - Madah Fatima
- Academic Department of Psychiatry and Behavioral Sciences, King Edward Medical University, Mayo Hospital, Lahore, Pakistan.
| |
Collapse
|
6
|
Brody BD, Park N, Christian A, Shaffer CW, Smetana R, Kotbi N, Russ MJ, Kanellopoulos D. Ketamine for major depressive disorder during an inpatient psychiatric admission: Effectiveness, adverse events, and lessons learned. J Affect Disord 2024; 351:293-298. [PMID: 38286230 DOI: 10.1016/j.jad.2024.01.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
OBJECTIVE Most studies examining the efficacy of ketamine for Major Depressive Disorder (MDD) have been conducted in outpatient or mixed inpatient/outpatient settings. Less is known about effectiveness and tolerability of ketamine for psychiatrically hospitalized patients. Efficacy and tolerability data from a naturalistic sample of acute inpatients may help inform institutions considering ketamine therapy for inpatient services. METHODS We performed a retrospective chart review of inpatients with non-psychotic MDD treated during the initial 3 years of a ketamine infusion program. Treatment effectiveness was defined using change in Montgomery Asberg Depression Rating Scale (MADRS) scores over five infusions. MDD treatment response was defined by a 50 % reduction of MADRS score, and remission was defined as MADRS score ≤ 10 at any point during the treatment. We also report the frequency of adverse events. RESULTS 41 patients with MDD were treated and had outcome data. 19 patients (46.5 %) met criteria for response and 15 patients (26.5 %) met criteria for remission during treatment. Four patients (10 %) had adverse psychological or behavioral outcomes. LIMITATIONS MADRS scales were administered by psychiatrists, psychologists, and trainees in each discipline who did not undergo standardized training in scale administration. Consistent data regarding the race/ethnicity of the patients was not available. CONCLUSION Twice weekly racemic ketamine infusion is an effective treatment option for patients hospitalized with MDD. Unmonitored or at home ketamine therapy may pose substantial risks.
Collapse
Affiliation(s)
- Benjamin D Brody
- Weill Cornell Medicine/Psychiatry, New York and White Plains, NY, United States of America.
| | - Nana Park
- New York Presbyterian Hospital, United States of America
| | | | - Charles W Shaffer
- Weill Cornell Medicine/Psychiatry, New York and White Plains, NY, United States of America
| | - Roy Smetana
- Weill Cornell Medicine/Psychiatry, New York and White Plains, NY, United States of America
| | - Nabil Kotbi
- Weill Cornell Medicine/Psychiatry, New York and White Plains, NY, United States of America
| | - Mark J Russ
- Silver Hill Hospital, United States of America
| | - Dora Kanellopoulos
- Weill Cornell Medicine/Psychiatry, New York and White Plains, NY, United States of America
| |
Collapse
|
7
|
Liu QR, Zong QK, Ding LL, Dai HY, Sun Y, Dong YY, Ren ZY, Hashimoto K, Yang JJ. Effects of perioperative use of esketamine on postpartum depression risk in patients undergoing cesarean section: A randomized controlled trial. J Affect Disord 2023; 339:815-822. [PMID: 37482224 DOI: 10.1016/j.jad.2023.07.103] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/27/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Postpartum depression (PPD) is a prevalent public health issue. Although ketamine has prophylactic effects on PPD in women undergoing cesarean section, the effects of esketamine on PPD remain unclear. This trial aimed to evaluate the efficacy of perioperative esketamine infusion on PPD risk by assessing Edinburgh Postnatal Depression Scale (EPDS) scores and blood biomarkers. METHODS A total of 150 participants undergoing elective cesarean section were randomly allocated to receive either esketamine or normal saline. Since 27 participants were excluded due to consent withdrawal or loss to follow-up, 123 patients were included. The primary outcome was the prevalence of PPD risk. Secondary outcomes included the prevalence of postpartum anxiety (PPA) risk, levels of biomarkers, postoperative pain intensity, and cumulative sufentanil consumption. RESULTS The prevalence of PPD and PPA risk at 3 days, 42 days, 3 months, and 6 months postpartum did not differ between the two groups. Furthermore, EPDS scores, pain intensity at rest, and during coughing on postoperative days (POD) 1 and 2 did not differ between the two groups. Sufentanil consumption during 0-12 h, 12-24 h, 0-24 h, and 0-48 h postoperatively were significantly lower in the esketamine group compared to the control group. Blood biomarkers did not differ between the two groups on POD 3. LIMITATIONS The sample size was small. PPD risk was simply screened, not diagnosed. CONCLUSIONS Perioperative administration of esketamine did not decrease the incidence of PPD risk in women after elective cesarean section. However, esketamine reduced opioid consumption.
Collapse
Affiliation(s)
- Qing-Ren Liu
- Department of Anesthesiology, Xishan People's Hospital of Wuxi City, Wuxi 214105, China
| | - Qian-Kun Zong
- Department of Anesthesiology, Xishan People's Hospital of Wuxi City, Wuxi 214105, China
| | - Li-Li Ding
- Department of Anesthesiology, Xishan People's Hospital of Wuxi City, Wuxi 214105, China
| | - Hong-Yan Dai
- Department of Obstetrics & Gynecology, Xishan People's Hospital of Wuxi City, Wuxi, 214105, China
| | - Yan Sun
- Department of Obstetrics & Gynecology, Xishan People's Hospital of Wuxi City, Wuxi, 214105, China
| | - Yong-Yan Dong
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Zhuo-Yu Ren
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
8
|
Gobira PH, LaMar J, Marques J, Sartim A, Silveira K, Santos L, Wegener G, Guimaraes FS, Mackie K, Lu HC, Joca S. CB1 Receptor Silencing Attenuates Ketamine-Induced Hyperlocomotion Without Compromising Its Antidepressant-Like Effects. Cannabis Cannabinoid Res 2023; 8:768-778. [PMID: 36067014 PMCID: PMC10771879 DOI: 10.1089/can.2022.0072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction: The antidepressant properties of ketamine have been extensively demonstrated in experimental and clinical settings. However, the psychotomimetic side effects still limit its wider use as an antidepressant. It was recently observed that endocannabinoids are inolved in ketamine induced reward properties. As an increase in endocannabinoid signaling induces antidepressant effects, this study aimed to investigate the involvement of cannabinoid type 1 receptors (CB1R) in the antidepressant and psychostimulant effects induced by ketamine. Methods: We tested the effects of genetic and pharmacological inhibition of CB1R in the hyperlocomotion and antidepressant-like properties of ketamine. The effects of ketamine (10-20 mg/kg) were assessed in the open-field and the forced swim tests (FSTs) in CB1R knockout (KO) and wild-type (WT) mice (male and female), and mice pre-treated with rimonabant (CB1R antagonist, 3-10 mg/kg). Results: We found that the motor hyperactivity elicited by ketamine was impaired in CB1R male and female KO mice. A similar effect was observed upon pharmacological blockade of CB1R in WT mice. However, genetic CB1R deletion did not modify the antidepressant effect of ketamine in male mice submitted to the FST. Surprisingly, pharmacological blockade of CB1R induced an antidepressant-like effect in both male and female mice, which was not further potentiated by ketamine. Conclusions: Our results support the hypothesis that CB1R mediate the psychostimulant side effects induced by ketamine, but not its antidepressant properties.
Collapse
Affiliation(s)
- Pedro Henrique Gobira
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Jacob LaMar
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA
| | - Jade Marques
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Ariandra Sartim
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Kennia Silveira
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Luana Santos
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | | | - Ken Mackie
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
- Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
- Program in Neuroscience, Indiana University, Bloomington, Indiana, USA
| | - Sâmia Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
9
|
Nikolin S, Rodgers A, Schwaab A, Bahji A, Zarate C, Vazquez G, Loo C. Ketamine for the treatment of major depression: a systematic review and meta-analysis. EClinicalMedicine 2023; 62:102127. [PMID: 37593223 PMCID: PMC10430179 DOI: 10.1016/j.eclinm.2023.102127] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/09/2023] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
Background Intranasal esketamine has received regulatory approvals for the treatment of depression. Recently a large trial of repeated dose racemic ketamine also demonstrated efficacy in severe depression. However, uncertainties remain regarding comparative efficacy, dosage, and the time course of response. Methods In this systematic review and meta-analysis, we searched Embase, Medline, Pubmed, PsycINFO, and CENTRAL up to April 13, 2023, for randomised controlled trials (RCTs) investigating ketamine for depression. Two investigators independently assessed study eligibility and risk of bias and extracted the data on depression severity scores, response and remission rates, and all-cause dropouts. Multivariable mixed-effects meta-regressions incorporated drug formulation (racemic (Rac) or esketamine (Esket)) and dose (Low or High) as covariates. Treatment effects were assessed: immediately following the first dose, during further repeated dosing, and follow-up after the final dose of a treatment course. This study is registered with PROSPERO (CRD42021221157). Findings The systematic review identified 687 articles, of which 49 RCTs were eligible for analysis, comprising 3299 participants. Standardised mean differences (95% confidence intervals) immediately following the first/single treatment were moderate-high for all conditions (Rac-High: -0.73, -0.91 to -0.56; Esket-High: -0.48, -0.75 to -0.20; Rac-Low: -0.33, -0.54 to -0.12; Esket-Low: -0.55, -0.87 to -0.24). Ongoing effects during repeated dosing were significantly greater than the control for Rac-High (-0.61; -1.02 to -0.20) and Rac-Low (-0.55, -1.09 to -0.00), but not Esket-Low (-0.15, -0.49 to 0.19) or Esket-High (-0.22, -0.54 to 0.10). At follow-up effects remained significant for racemic ketamine (-0.65; -1.23 to -0.07) but not esketamine (-0.33; -0.96 to 0.31). All-cause dropout was similar between experiment and control conditions for both formulations combined (Odds Ratio = 1.18, 0.85-1.64). Overall heterogeneity varied from 5.7% to 87.6. Interpretation Our findings suggested that effect sizes for depression severity, as well as response and remission rates, were numerically greater for racemic ketamine than esketamine. Higher doses were more effective than low doses. Differences were evident in initial effects, ongoing treatment, and lasting effects after the final dose. Funding None.
Collapse
Affiliation(s)
- Stevan Nikolin
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, Australia
- Black Dog Institute, Sydney, Australia
| | - Anthony Rodgers
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | | | - Anees Bahji
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carlos Zarate
- Section Neurobiology and Treatment of Mood Disorders, Division of Intramural Research Program, National Institute of Mental Health, 10 Center Drive, MSC 1282, Building 10CRC, Room 7-5342, Bethesda, MD 20892, USA
| | - Gustavo Vazquez
- Section Neurobiology and Treatment of Mood Disorders, Division of Intramural Research Program, National Institute of Mental Health, 10 Center Drive, MSC 1282, Building 10CRC, Room 7-5342, Bethesda, MD 20892, USA
| | - Colleen Loo
- Discipline of Psychiatry and Mental Health, University of New South Wales, Sydney, Australia
- Black Dog Institute, Sydney, Australia
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
10
|
Oyetunji A, Huelga C, Bunte K, Tao R, Bellman V. Use of ketamine for depression and suicidality in cancer and terminal patients: Review of current data. AIMS Public Health 2023; 10:610-626. [PMID: 37842268 PMCID: PMC10567968 DOI: 10.3934/publichealth.2023043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/17/2023] Open
Abstract
Depression and suicidality are significant challenges faced by cancer patients, particularly those in advanced stages of the disease or nearing the end of life. Conventional antidepressant therapies often have limited effectiveness or delayed onset of action, making the exploration of alternative treatments crucial. The use of ketamine as a potential treatment for depression and suicidality in cancer and terminal patients has gained considerable attention in recent years. This review article aims to provide a comprehensive analysis of the current data regarding the efficacy and safety of ketamine in this specific population. This review presents an overview of clinical trials and case studies investigating the use of ketamine in this population. It explores the effectiveness of ketamine as a standalone treatment or in combination with other interventions. Furthermore, the article addresses the limitations and future directions of research in this field. It highlights the need for larger, well-controlled studies with long-term follow-up to establish the efficacy, safety and optimal treatment parameters of ketamine for depression and suicidality in palliative care.
Collapse
Affiliation(s)
- Aderonke Oyetunji
- University of Missouri Kansas City, Psychiatry Residency Training Program, Kansas City, MO
| | - Christian Huelga
- Kansas City University, College of Osteopathic Medicine, Kansas City, MO
| | - Kailee Bunte
- University of Missouri Kansas City, School of Medicine, Kansas City, MO
| | - Rachel Tao
- University of Missouri Kansas City, School of Medicine, Kansas City, MO
| | - Val Bellman
- University of Missouri Kansas City, Psychiatry Residency Training Program, Kansas City, MO
| |
Collapse
|
11
|
Gargori NMB, Alahwal H, Vacheron CH, Alam FMA. Predictive factors of success and failure for intravenous ketamine therapy in patients suffering from chronic neuropathic pain. Saudi J Anaesth 2023; 17:340-348. [PMID: 37601528 PMCID: PMC10435804 DOI: 10.4103/sja.sja_737_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/06/2022] [Indexed: 08/22/2023] Open
Abstract
Background Intravenous (IV) ketamine is used for chronic neuropathic pain refractory to other treatments. Administration of such a medication requires high-cost services while the result is not always satisfactory with a significant percentage of failure. Success and failure are related to some factors. Method In this study, we aim to point the most responsive disease category to IV ketamine and the predictive factors for successful and failed treatment. Two hundred and sixty-nine patients out of 371 were included. Demographic, clinical, and therapy-related variables were retrospectively collected and then statistically analyzed using various descriptive and inferential methods. A few descriptive statistics are obtained for the variables depending on their nature (e.g., percentages for qualitative variables and means for quantitative variables). Furthermore, several inferential methods are considered to address some statistical points of interest, including, but not limited to, odds ratio interpretations via logistic regression as well as association and correlation analyses. Results A significant association was found between the presence of intermittent pain pattern as well as the pain of chronic primary category and favorable response to IV ketamine while the history of previous analgesic interventions was significantly associated with a negative response. Conclusion The identified factors can be used to prospectively study the efficacy of ketamine using selection criteria based on the observed results in our study to re-evaluate the percentage of responsiveness according to these new parameters.
Collapse
Affiliation(s)
- Nezar M. B. Gargori
- Department of Anesthesia and Intensive Care, Faculty of Medicine, Abdulaziz University, Jeddah, Western Region, Kingdom of Saudi Arabia
| | - Hazem Alahwal
- Department of Anesthesia and Intensive Care, Faculty of Medicine, Abdulaziz University, Jeddah, Western Region, Kingdom of Saudi Arabia
| | | | - Farouq Mohammad A. Alam
- Department of Statistics,Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Kim A, Gu SM, Lee H, Kim DE, Hong JT, Yun J, Cha HJ. Prenatal ketamine exposure impairs prepulse inhibition via arginine vasopressin receptor 1A-mediated GABAergic neuronal dysfunction in the striatum. Biomed Pharmacother 2023; 160:114318. [PMID: 36738499 DOI: 10.1016/j.biopha.2023.114318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Ketamine is a widely used anesthetic with N-methyl-D-aspartate (NMDA) receptor antagonism. Exposure to ketamine and NMDA receptor antagonists may induce psychosis. However, the mechanism underlying the effects of ketamine on the immature brain remains unclear. In this study, NMDA receptor antagonists, ketamine and methoxetamine, were administered to pregnant F344 rats (E17). These regimens induce psychosis-like behaviors in the offspring, such as hyperlocomotion induced by MK-801, a non-competitive NMDA receptor antagonist. We also observed that prepulse inhibition (PPI) was significantly reduced. Interestingly, ketamine administration increased the arginine vasopressin receptor 1A (Avpr1a) expression levels in the striatum of offspring with abnormal behaviors. Methoxetamine, another NMDA receptor antagonist, also showed similar results. In addition, we demonstrated a viral vector-induced Avpr1a overexpression in the striatum-inhibited PPI. In the striatum of offspring, ketamine or methoxetamine treatment increased glutamate decarboxylase 67 (GAD67) and δ-aminobutyric acid (GABA) levels. These results show that prenatal NMDA receptor antagonist treatment induces GABAergic neuronal dysfunction and abnormalities in sensorimotor gating via regulating Avpr1a expression in the striatum.
Collapse
Affiliation(s)
- Aeseul Kim
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Sun Mi Gu
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Haemiru Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Dong Eun Kim
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea.
| | - Hye Jin Cha
- College of Veterinary Medicine, Gyeongsang National University, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do 52828, the Republic of Korea.
| |
Collapse
|
13
|
Tian F, Lewis LD, Zhou DW, Balanza GA, Paulk AC, Zelmann R, Peled N, Soper D, Santa Cruz Mercado LA, Peterfreund RA, Aglio LS, Eskandar EN, Cosgrove GR, Williams ZM, Richardson RM, Brown EN, Akeju O, Cash SS, Purdon PL. Characterizing brain dynamics during ketamine-induced dissociation and subsequent interactions with propofol using human intracranial neurophysiology. Nat Commun 2023; 14:1748. [PMID: 36991011 PMCID: PMC10060225 DOI: 10.1038/s41467-023-37463-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Ketamine produces antidepressant effects in patients with treatment-resistant depression, but its usefulness is limited by its psychotropic side effects. Ketamine is thought to act via NMDA receptors and HCN1 channels to produce brain oscillations that are related to these effects. Using human intracranial recordings, we found that ketamine produces gamma oscillations in prefrontal cortex and hippocampus, structures previously implicated in ketamine's antidepressant effects, and a 3 Hz oscillation in posteromedial cortex, previously proposed as a mechanism for its dissociative effects. We analyzed oscillatory changes after subsequent propofol administration, whose GABAergic activity antagonizes ketamine's NMDA-mediated disinhibition, alongside a shared HCN1 inhibitory effect, to identify dynamics attributable to NMDA-mediated disinhibition versus HCN1 inhibition. Our results suggest that ketamine engages different neural circuits in distinct frequency-dependent patterns of activity to produce its antidepressant and dissociative sensory effects. These insights may help guide the development of brain dynamic biomarkers and novel therapeutics for depression.
Collapse
Affiliation(s)
- Fangyun Tian
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura D Lewis
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Radiology, MGH/HST Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
- Institute for Medical Engineering and Sciences, Department of Electrical Engineering and Computer Science, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David W Zhou
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gustavo A Balanza
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Angelique C Paulk
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
| | - Rina Zelmann
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
| | - Noam Peled
- Department of Radiology, MGH/HST Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA, USA
| | - Daniel Soper
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura A Santa Cruz Mercado
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert A Peterfreund
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Linda S Aglio
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Emad N Eskandar
- Department of Neurological Surgery, Albert Einstein College of Medicine, Bronx, NY, USA
| | - G Rees Cosgrove
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Ziv M Williams
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emery N Brown
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sydney S Cash
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick L Purdon
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Sachdeva B, Sachdeva P, Ghosh S, Ahmad F, Sinha JK. Ketamine as a therapeutic agent in major depressive disorder and posttraumatic stress disorder: Potential medicinal and deleterious effects. IBRAIN 2023; 9:90-101. [PMID: 37786516 PMCID: PMC10528797 DOI: 10.1002/ibra.12094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 10/04/2023]
Abstract
Major depressive disorder (MDD) and posttraumatic stress disorder (PTSD) are the most common causes of emotional distress that impair an individual's quality of life. MDD is a chronic mental illness that affects 300 million people across the world. Clinical manifestations of MDD include fatigue, loss of interest in routine tasks, psychomotor agitation, impaired ability to focus, suicidal ideation, hypersomnolence, altered psychosocial functioning, and appetite loss. Individuals with depression also demonstrate a reduced behavioral response while experiencing pleasure, a symptom known as anhedonia. Like MDD, PTSD is a prevalent and debilitating psychiatric disorder resulting from a traumatic incident such as sexual assault, war, severe accident, or natural disaster. Symptoms such as recalling event phases, hypervigilance, irritability, and anhedonia are common in PTSD. Both MDD and PTSD pose enormous socioeconomic burdens across the globe. The search for effective treatment with minimal side effects is still ongoing. Ketamine is known for its anesthetic and analgesic properties. Psychedelic and psychotropic effects of ketamine have been found on the nervous system, which highlights its toxicity. In this article, the effectiveness of ketamine as a potential therapeutic for PTSD and MDD along with its mechanisms of action, clinical trials, and possible side effects have been discussed.
Collapse
Affiliation(s)
- Bhuvi Sachdeva
- Department of Physics and Astrophysics, Bhagini Nivedita CollegeUniversity of DelhiDelhiIndia
| | | | - Shampa Ghosh
- GloNeuro AcademyNoidaUttar PradeshIndia
- ICMR—National Institute of NutritionTarnakaHyderabadIndia
| | - Faizan Ahmad
- Department of Medical Elementology and ToxicologyJamia HamdardDelhiIndia
| | | |
Collapse
|
15
|
Głuch-Lutwin M, Sałaciak K, Pytka K, Gawalska A, Jamrozik M, Śniecikowska J, Kołaczkowski M, Depoortère RY, Newman-Tancredi A. The 5-HT 1A receptor biased agonist, NLX-204, shows rapid-acting antidepressant-like properties and neurochemical changes in two mouse models of depression. Behav Brain Res 2023; 438:114207. [PMID: 36368443 DOI: 10.1016/j.bbr.2022.114207] [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: 09/20/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
Abstract
Activation of cortical serotonin 5-HT1A receptors may be a promising strategy to achieve rapid-acting antidepressant (RAAD) activity. NLX-204 is a selective 5-HT1A receptor biased agonist that, in naïve mice, robustly decreases immobility in the forced swim test (FST), and preferentially phosphorylates extracellular signal-regulated kinase (ERK1/2), involved in antidepressant activity. Here, we evaluated the properties of NLX-204 in two mouse models of depression. Male CD-1 mice were subjected to unpredictable chronic mild stress (UCMS) for 4-weeks or to repeated corticosterone (CORT, 20 mg/kg s.c./day) for 3-weeks before receiving acute administration of NLX-204 (2 mg/kg, p.o.). Depressive-like behavior was assessed in the FST, anhedonia-like behavior in the sucrose preference test (SPT) and locomotor activity was also recorded. Phosphorylation of ERK1/2 (pERK1/2) and cAMP response binding element (pCREB) were measured ex vivo in hippocampus and prefrontal cortex (PFC). UCMS or CORT treatment increased immobility in the FST, elicited a sucrose preference deficit, and decreased pERK1/2 and pCREB levels in PFC and hippocampus. NLX-204 reduced depressive-like behavior in the FST in CORT and UCMS mice, and normalized sucrose preference in CORT mice, suggesting anti-anhedonic activity. NLX-204 increased pERK1/2 levels in PFC of UCMS mice. NLX-204 also increased pCREB levels in PFC of CORT mice. These data suggest that NLX-204 has RAAD-like properties not only in naïve mice, but also in mice in a "depressive-like" state, and that these involve changes in PFC and hippocampal pERK1/2 and pCREB levels. These data provide additional evidence that activation of 5-HT1A receptors by selective biased agonists, such as NLX-204, may constitute a promising RAAD strategy.
Collapse
Affiliation(s)
- Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Alicja Gawalska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Marek Jamrozik
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Śniecikowska
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Marcin Kołaczkowski
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | | | | |
Collapse
|
16
|
Smith-Apeldoorn SY, Veraart JK, Spijker J, Kamphuis J, Schoevers RA. Maintenance ketamine treatment for depression: a systematic review of efficacy, safety, and tolerability. Lancet Psychiatry 2022; 9:907-921. [PMID: 36244360 DOI: 10.1016/s2215-0366(22)00317-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/30/2022]
Abstract
Ketamine has rapid yet often transient antidepressant effects in patients with treatment-resistant depression. Different strategies have been proposed to prolong these effects. Maintenance ketamine treatment appears promising, but little is known about its efficacy, safety, and tolerability in depression. We searched Pubmed, Embase, and the Cochrane Library and identified three randomised controlled trials, eight open-label trials, and 30 case series and reports on maintenance ketamine treatment. We found intravenous, intranasal, oral, and possibly intramuscular and subcutaneous maintenance ketamine treatment to be effective in sustaining antidepressant effect in treatment-resistant depression. Tachyphylaxis, cognitive impairment, addiction, and serious renal and urinary problems seem uncommon. Despite the methodological limitations, we conclude that from a clinical view, maintenance ketamine treatment seems to be of therapeutic potential. We recommend both controlled and naturalistic studies with long-term follow-up and sufficient power to determine the position of maintenance ketamine treatment within routine clinical practice.
Collapse
Affiliation(s)
- Sanne Y Smith-Apeldoorn
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
| | - Jolien Ke Veraart
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, Netherlands; Department of Mood Disorders, PsyQ Haaglanden, Parnassia Psychiatric Institute, The Hague, Netherlands
| | - Jan Spijker
- Pro Persona Mental Health Care, Depression Expertise Center, Nijmegen, Netherlands
| | - Jeanine Kamphuis
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Robert A Schoevers
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
17
|
Shamabadi A, Ahmadzade A, Hasanzadeh A. Ketamine for suicidality: An umbrella review. Br J Clin Pharmacol 2022; 88:3990-4018. [PMID: 35451097 DOI: 10.1111/bcp.15360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 12/01/2022] Open
Abstract
The urgent need for appropriate treatment for suicide, the tenth leading cause of death, has led to numerous studies. This study aims to systematically identify and appraise systematic reviews with or without meta-analyses investigating ketamine in suicidal ideation and behaviours. Scopus, ISI, Embase, PubMed, CINAHL, PsycINFO, Cochrane Library, Google Scholar and two registries were searched without any restrictions for systematic reviews investigating the efficacy of ketamine on suicidal ideation and behaviours. The primary outcome was the final inference of ketamine effectiveness. A formal narrative synthesis was conducted, and the AMSTAR-2 tool was used to evaluate the quality of the studies. Of 27 studies that addressed ketamine for suicidal ideation, only four reported mixed or negative results, and out of nine reviews, esketamine was significantly beneficial only in five. A transient rise in pulse rate and blood pressure, dissociation, confusion, blurred vision, nausea and vertigo were the most common adverse effects; however, most were mild. More than two-thirds of the included studies qualified as of low or critically low quality. Preliminary evidence for the short-term efficacy of ketamine in suicidality was noted by the majority of reviews; however, long-term effects remained unknown. Due to the low quality of many studies and the limitations of core studies, further research is required.
Collapse
Affiliation(s)
- Ahmad Shamabadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadzade
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Hasanzadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Ren L, Zhang H, Tao W, Chen Y, Zou Z, Guo X, Shen Q, Feng Q, Hu J. The Rapid and Long-Lasting Antidepressant Effects of Iridoid Fraction in Gardenia Jasminoides J.Ellis Are Dependent on Activating PKA-CREB Signaling Pathway. Front Pharmacol 2022; 13:896628. [PMID: 35754496 PMCID: PMC9213885 DOI: 10.3389/fphar.2022.896628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Lag periods of therapeutic efficacy cause poor compliance of patients, which has made solutions for rapid antidepressants the most urgent need in the depression study field at present. We have identified through our previous studies the rapid antidepressant effects of the traditional herb Gardenia jasminoides J.Ellis [Rubiaceae] (GJ) and its standardized fractions. Through screening different fractions of GJ, we decided to place our focus on the iridoid fraction of GJ (GJ-IF). Methods: 1. Tail suspension test (TST), forced swimming test (FST), and novelty suppressed-feeding test (NSFT) were performed in sequence on mice after GJ-IF administration. 2. Mice in the model group were under chronic unpredictable mild stress (CUMS) for 3 w. After GJ-IF treatment, mice were placed in an open field test (OFT), Sucrose preference test (SPT), NSFT, TST, and FST. 3. Western Blot was performed to examine the expression of brain-derived neurotrophic factor (BDNF), Synapsin 1, cyclic-AMP dependent protein kinase A (PKA), phosphorylated cyclic-AMP responsive element-binding protein (p-CREB), and cAMP response element-binding protein (CREB). 4. Mice in the test group were administrated with GJ-IF after intraperitoneal injection of PKA blocker H89. Results: 1. GJ-IF treatment significantly reduced the immobility time of TST at 1 d and FST at 26 h. 2. GJ-IF reversed the deficits induced by 3 w CUMS in SPT, TST, FST, and NSFT at 1 d and 26 h. The antidepressant effects of a single dose of iridoid fraction could also last for at least 14 d. 3. The results of molecule studies suggested that a single dose of GJ-IF activated p-CREB at 2 h and the PKA-CREB pathway at 1 d. The expression of BDNF did not significantly change from 30 min to 1 d after GJ-IF administration. 4. Blockade of PKA-CREB signaling pathway reversed the antidepressant effects of GJ-IF at 1 d, but not 30 min and 2 h. Conclusion: GJ-IF is the crucial component in the rapid antidepressant of GJ. Rapid and sustained antidepressant effects of GJ-IF were dependent on activating the PKA-CREB signaling pathway.
Collapse
Affiliation(s)
- Li Ren
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders and School of Chinese Medicine, Jinan University, Guangzhou, China
| | - Weiwei Tao
- Basic Teaching and Research Department of Integrated Chinese and Western Medicine, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yin Chen
- Basic Teaching and Research Department of Integrated Chinese and Western Medicine, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhilu Zou
- Basic Teaching and Research Department of Integrated Chinese and Western Medicine, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - XiaoYan Guo
- Basic Teaching and Research Department of Integrated Chinese and Western Medicine, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qinqin Shen
- Basic Teaching and Research Department of Integrated Chinese and Western Medicine, College of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Quansheng Feng
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingqing Hu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
19
|
Zhang H, Sun Y, Yau SY, Zhou Y, Song X, Zhang HT, Zhu B, Wu H, Chen G. Synergistic effects of two natural compounds of iridoids on rapid antidepressant action by up-regulating hippocampal PACAP signaling. Br J Pharmacol 2022; 179:4078-4091. [PMID: 35362097 DOI: 10.1111/bph.15847] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/30/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Current mainstream antidepressants have limited efficacy and a delayed onset of action. Yueju is a traditional herbal medicine conferring rapid antidepressant activity. Here we attempted to identify the effective compounds from Yueju and the underlying mechanisms. EXPERIMENTAL APPROACH A transcriptomic analysis was employed to discover key candidate molecules for rapid antidepressant response. The enriched compounds in Yueju were identified with HPLC. Antidepressant effects were evaluated periodically using various behavioral paradigms. The mechanistic signaling was assessed using site-directed pharmacological intervention or optogenetic manipulation. KEY RESULTS A transcriptomic analysis revealed that Yueju up-regulated pituitary adenylate cyclase activating polypeptide (PACAP) expression in the hippocampus. Two iridoids geniposide (GP) and shanzhiside methyl-ester (SM) were enriched in Yueju. Co-treatment of GP and SM each at an equivalent dose in Yueju synergistically increased PACAP expression and elicited rapid antidepressant effects, which were prevented by intra-hippocampal dentate gyrus (DG) infusions of a PACAP antagonist or optogenetic inactivation of PACAP-expressing neurons. GP-SM co-treatment rapidly reduced CaMKII phosphorylation and enhanced mTOR/4EBP1/P70S6k/BDNF signaling, while intra-DG infusions of a CaMKII activator blunted rapid antidepressant effects and BDNF expression up-regulation induced by GP-SM co-treatment. A single administration of GP-SM rapidly improved depression-like behaviors and up-regulated hippocampal PACAP signaling in the repeated corticosterone-induced depression model, further confirming its rapid antidepressant action and the involvement of PACAP. CONCLUSION AND IMPLICATIONS GP-SM co-treatment elicited a synergistic effect on rapid antidepressant effects via triggering hippocampal PACAP activity and associated CaMKII-BDNF signaling, shedding lights on the development of novel targeted antidepressants.
Collapse
Affiliation(s)
- Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders & School of Chinese Medicine, Jinan University, Guangzhou, China.,College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Co-innovation Center of Neurogeneration, Nantong University, Nantong, Jiangsu, China
| | - Yan Sun
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders & School of Chinese Medicine, Jinan University, Guangzhou, China.,College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China
| | - Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Xinxin Song
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, the Rockefeller Neuroscience Institute, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Boran Zhu
- College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoxin Wu
- College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Gang Chen
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders & School of Chinese Medicine, Jinan University, Guangzhou, China.,Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, China.,Co-innovation Center of Neurogeneration, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
20
|
Giri AR, Kaur N, Yarrarapu SNS, Rottman Pietrzak KA, Santos C, Lowman PE, Niaz S, Franco PM, Sanghavi DK. "Novel Management of Depression Using Ketamine in the Intensive Care Unit". J Intensive Care Med 2022; 37:1654-1661. [PMID: 35313768 DOI: 10.1177/08850666221088220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ketamine, a dissociative anesthetic, induces improvement in depressive symptoms by antagonizing glutaminergic NMDA receptors. Ketamine has been used previously in outpatient setting for treatment-resistant depression, but we showcase its utility in depression management at the Intensive Care Unit (ICU). Research Question: Can ketamine be used for depression treatment in ICU patients? Study Design and Methods: A retrospective chart review of ICU patients was done at a tertiary center from 2018 to 2021, to assess the ketamine usage. Among the patients reviewed, ketamine was used for depression in 12, and for analgesia & sedation in 2322 patients. Ketamine was administered in doses of 0.5mg/kg & 0.75mg/kg for depression. Each course consisted of 3 doses of ketamine administered over 3 days, and 7 in 12 patients received a single course of ketamine. The rest received 3-4 courses 1 week apart. Results: Ketamine was found to improve mood and affect in most of the patients with depression. 11 in 12 patients had a positive response with better sleep. It has a major advantage over conventional anti-depressants since it takes only a few hours to induce clinical improvement. Patients who were observably withdrawn from care team and family, were administered ketamine. Conclusion: A major drawback of ketamine is that the duration of clinical improvement is short, with the response lasting only up to seven days after a single dose. Hence, all the patients in our study were weaned off ketamine with a supporting antidepressant. Ketamine has been documented to cause cardio-neurotoxicity; however, only one patient had worsening lethargy in our study. To conclude, ketamine has a marked benefit in treating depression in the ICU. Although our study was associated with positive outcomes, there is a need for prospective studies with long-term follow-up assessments.
Collapse
|
21
|
Subramanian S, Haroutounian S, Palanca BJA, Lenze EJ. Ketamine as a therapeutic agent for depression and pain: mechanisms and evidence. J Neurol Sci 2022; 434:120152. [PMID: 35092901 DOI: 10.1016/j.jns.2022.120152] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/14/2022]
Abstract
Ketamine is an anesthetic drug which is now used to treat chronic pain conditions and psychiatric disorders, especially depression. It is an N-methyl-D-aspartate (NMDA) receptor antagonist with additional effects on α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, opioid receptors, and monoaminergic receptors. This article focuses on ketamine's role in treating depression and pain, two commonly comorbid challenging conditions with potentially shared neurobiologic circuitry. Many clinical trials have utilized intravenous or intranasal ketamine for treating depression and pain. Intravenous ketamine is more bioavailable than intranasal ketamine and both are effective for acute depressive episodes. Intravenous ketamine is advantageous for post-operative analgesia and is associated with a reduction in total opioid requirements. Few studies have treated chronic pain or concurrent depression and pain with ketamine. Larger, randomized control trials are needed to examine the safety and efficacy of intravenous vs. intranasal ketamine, ideal target populations, and optimal dosing to treat both depression and pain.
Collapse
Affiliation(s)
- Subha Subramanian
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ben Julian A Palanca
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric J Lenze
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
22
|
Palamar JJ, Kumar S, Yang KH, Han BH. Ketamine use in relation to depressive symptoms among high school seniors. Am J Addict 2022; 31:100-107. [PMID: 35076151 PMCID: PMC8901529 DOI: 10.1111/ajad.13259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/29/2021] [Accepted: 01/05/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Ketamine is efficacious in treating treatment-resistant depression in medical settings and the drug was approved for such use by the US Federal Drug Administration in 2019. However, little is known about how use outside of medical settings relates to depression. We determined whether recreational ketamine use, relative to the use of other drugs, is related to the current experience of depression among adolescents. METHODS We examined data from the 2016 to 2019 Monitoring the Future nationally representative survey of high school seniors in the United States (N = 15,673). We determined how past-year drug use and frequency of past-year drug use were associated with students reporting a high level of current depressive symptoms relative to other students. RESULTS Ketamine use was associated with highest risk for a high level of depression (aPR = 1.55, 95% confidence interval [CI]: 1.24-1.94), followed by use of cannabis (aPR = 1.29, 95% CI: 1.19-1.39), and nonmedical use of tranquilizers (aPR = 1.22, 95% CI: 1.04-1.44) and amphetamine (aPR = 1.17, 95% CI: 1.01-1.34). Alcohol use was associated with decreased risk (aPR = 0.92, 95% CI: 0.85-0.99). With respect to frequency of past-year use, more frequent use of ketamine and cannabis was associated with increased risk for a high level of depression in a dose-response-like manner, with past-year use of ketamine and cannabis ≥10 times associated with increased risk for depression by 70% and 40%, respectively. DISCUSSION AND CONCLUSIONS Past-year recreational ketamine use is a risk factor for reporting current depression than most other drugs. SCIENTIFIC SIGNIFICANCE This was the first study to compare the risk of use of various drugs in relation to depression.
Collapse
Affiliation(s)
- Joseph J. Palamar
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| | - Sakthi Kumar
- School of Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Kevin H. Yang
- Department of Medicine, Division of Geriatrics, Gerontology, and Palliative Care, University of California San Diego, San Diego, California, USA
| | - Benjamin H. Han
- Department of Medicine, Division of Geriatrics, Gerontology, and Palliative Care, University of California San Diego, San Diego, California, USA
| |
Collapse
|
23
|
Scotton E, Antqueviezc B, Vasconcelos M, Dalpiaz G, Paul Géa L, Ferraz Goularte J, Colombo R, Ribeiro Rosa A. Is (R)-ketamine a Potential Therapeutic Agent for Treatment-Resistant Depression with Less Detrimental Side Effects? A Review of Molecular Mechanisms Underlying Ketamine and its Enantiomers. Biochem Pharmacol 2022; 198:114963. [PMID: 35182519 DOI: 10.1016/j.bcp.2022.114963] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 12/18/2022]
Abstract
Approximately one-third of individuals with major depressive disorder are resistant to conventional antidepressants (i.e., monoamine-based therapies), and, even among respondents, a proper therapeutic effect may require weeks of treatment. Ketamine, a racemic mixture of the two enantiomers, (R)-ketamine and (S)-ketamine, is an N-methyl-d-aspartate receptor (NMDAR) antagonist and has been shown to have rapid-acting antidepressant properties in patients with treatment-resistant depression (TRD). Although (R)-ketamine has a lower affinity for NMDAR, it presents greater potency and longer-lasting antidepressant properties, with no major side effects, than racemic ketamine or (S)-ketamine in preclinical findings. Thereby, ketamine and its enantiomers have not only an antagonistic effect on NMDAR but also a strong synaptogenic-modulatory effect, which is impaired in TRD pathophysiology. In this review, we summarize the current evidence regarding the modulation of neurotransmission, neuroplasticity, and neural network activity as putative mechanisms of these rapid-acting antidepressants, highlighting differences on intracellular signaling pathways of synaptic proteins such as mammalian target of rapamycin (mTOR), extracellular signal-regulated kinase (ERK) and brain-derived neurotrophic factor (BDNF). In addition, we discuss probable mechanisms involved in the side effects of ketamine and its enantiomers.
Collapse
Affiliation(s)
- Ellen Scotton
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Pharmacology, Programa de Pós-Graduação em Farmacologia e Terapêutica, UFRGS, Porto Alegre, RS, Brazil.
| | - Bárbara Antqueviezc
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Mailton Vasconcelos
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Instituto de Psicologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Giovana Dalpiaz
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Luiza Paul Géa
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| | - Jéferson Ferraz Goularte
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Rafael Colombo
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Biotecnologia, Universidade de Caxias do Sul (UCS), Caxias do Sul, RS, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade de Caxias do Sul (UCS), Caxias do Sul, RS, Brazil.
| | - Adriane Ribeiro Rosa
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Department of Pharmacology, Programa de Pós-Graduação em Farmacologia e Terapêutica, UFRGS, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
24
|
Gastaldon C, Laurenzi PF, Schoretsanitis G, Papola D, Cristea IA, Naudet F, Ostuzzi G, Barbui C. Esketamine for treatment-resistant depression in adults. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2022. [DOI: 10.1002/14651858.cd015071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Chiara Gastaldon
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry; University of Verona; Verona Italy
- Cochrane Global Mental Health; University of Verona; Verona Italy
| | | | - Georgios Schoretsanitis
- Department of Psychiatry; The Zucker Hillside Hospital; Northwell Health, Glen Oaks New York USA
- Department of Psychiatry; Zucker School of Medicine at Northwell/Hofstra; Hempstead New York USA
- Department of Psychiatry, Psychotherapy and Psychosomatics, Hospital of Psychiatry; University of Zurich; Zurich Switzerland
| | - Davide Papola
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry; University of Verona; Verona Italy
- Cochrane Global Mental Health; University of Verona; Verona Italy
| | - Ioana Alina Cristea
- Department of Brain and Behavioral Sciences; University of Pavia; Pavia Italy
| | - Florian Naudet
- University Rennes, CHU Rennes, Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes); Rennes France
| | - Giovanni Ostuzzi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry; University of Verona; Verona Italy
| | - Corrado Barbui
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry; University of Verona; Verona Italy
- Cochrane Global Mental Health; University of Verona; Verona Italy
| |
Collapse
|
25
|
Goswamee P, Rice R, Leggett E, Zhang F, Manicka S, Porter JH, McQuiston AR. Effects of subanesthetic ketamine and (2R,6R) hydroxynorketamine on working memory and synaptic transmission in the nucleus reuniens in mice. Neuropharmacology 2022; 208:108965. [PMID: 35065945 PMCID: PMC8885971 DOI: 10.1016/j.neuropharm.2022.108965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 01/07/2022] [Accepted: 01/15/2022] [Indexed: 01/16/2023]
Abstract
RATIONALE Acute cognitive impairment and abuse potential of ketamine incentivizes the search for alternatives to ketamine for clinical management of treatment-resistant depression. Recently, (2R,6R) hydroxynorketamine ((2R,6R)-HNK), a metabolite of ketamine, has shown promise due to its reported lack of ketamine-like reinforcing properties. Nonetheless, the effect of (2R,6R)-HNK on cognition has not been reported. METHOD Adult male mice were placed in a Y-maze to measure spatial working memory (SWM) 24 h after treatment with either a single or repeated subanesthetic dose of (2R,6R)-HNK or ketamine. To determine the effect of the drug regimens on synaptic mechanisms in neural circuits deemed critical for SWM, we conducted patch-clamp electrophysiological recordings from neurons in the midline thalamic nucleus reuniens (RE) in response to optogenetic stimulation of medial prefrontal cortex (mPFC) inputs in acutely prepared brain slices. RESULTS Single or repeated treatment with a 10 mg/kg dose of either drug did not impact performance in a Y-maze. However, single administration of a ½-log higher dose (32 mg/kg) of ketamine significantly reduced SWM. The same dose of (2R,6R)-HNK did not produce SWM deficits. Interestingly, repeated administration of either drugs at the 32 mg/kg had no effect on SWM performances. Concomitant to these effects on SWM, only single injection of 32 mg/kg of ketamine was found to increase the mPFC-driven action potential firing activity in the RE neurons. Conversely, both single and repeated administration of the 32 mg/kg dose of (2R,6R)-HNK but not ketamine, increased the input resistance of the RE neurons. CONCLUSION Our results indicate that acute treatment of ketamine at 32 mg/kg increases mPFC-driven firing activity of RE neurons, and this contributes to the ketamine-mediated cognitive deficit. Secondly, sub-chronic treatment with the same dose of ketamine likely induces tolerance. Although single or repeated administration of the 32 mg/kg dose of (2R,6R)-HNK can alter intrinsic properties of RE neurons, this dose does not produce cognitive deficit or changes in synaptic mechanism in the RE.
Collapse
Affiliation(s)
- Priyodarshan Goswamee
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Remington Rice
- Department of Psychology, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Elizabeth Leggett
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Fan Zhang
- Department of Psychology, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Sofia Manicka
- Department of Psychology, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Joseph H Porter
- Department of Psychology, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - A Rory McQuiston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
26
|
Riggs LM, An X, Pereira EFR, Gould TD. (R,S)-ketamine and (2R,6R)-hydroxynorketamine differentially affect memory as a function of dosing frequency. Transl Psychiatry 2021; 11:583. [PMID: 34772915 PMCID: PMC8590048 DOI: 10.1038/s41398-021-01685-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
A single subanesthetic infusion of ketamine can rapidly alleviate symptoms of treatment-resistant major depression. Since repeated administration is required to sustain symptom remission, it is important to characterize the potential untoward effects of prolonged ketamine exposure. While studies suggest that ketamine can alter cognitive function, it is unclear to what extent these effects are modulated by the frequency or chronicity of treatment. To test this, male and female adolescent (postnatal day [PD] 35) and adult (PD 60) BALB/c mice were treated for four consecutive weeks, either daily or thrice-weekly, with (R,S)-ketamine (30 mg/kg, intraperitoneal) or its biologically active metabolite, (2R,6R)-hydroxynorketamine (HNK; 30 mg/kg, intraperitoneal). Following drug cessation, memory performance was assessed in three operationally distinct tasks: (1) novel object recognition to assess explicit memory, (2) Y-maze to assess working memory, and (3) passive avoidance to assess implicit memory. While drug exposure did not influence working memory performance, thrice-weekly ketamine and daily (2R,6R)-HNK led to explicit memory impairment in novel object recognition independent of sex or age of exposure. Daily (2R,6R)-HNK impaired implicit memory in the passive-avoidance task whereas thrice-weekly (2R,6R)-HNK tended to improve it. These differential effects on explicit and implicit memory possibly reflect the unique mechanisms by which ketamine and (2R,6R)-HNK alter the functional integrity of neural circuits that subserve these distinct cognitive domains, a topic of clinical and mechanistic relevance to their antidepressant actions. Our findings also provide additional support for the importance of dosing frequency in establishing the cognitive effects of repeated ketamine exposure.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Xiaoxian An
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Edna F R Pereira
- Department of Epidemiology and Public Health, Division of Translational Toxicology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA.
| |
Collapse
|
27
|
Karrouri R, Hammani Z, Benjelloun R, Otheman Y. Major depressive disorder: Validated treatments and future challenges. World J Clin Cases 2021; 9:9350-9367. [PMID: 34877271 PMCID: PMC8610877 DOI: 10.12998/wjcc.v9.i31.9350] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
Depression is a prevalent psychiatric disorder that often leads to poor quality of life and impaired functioning. Treatment during the acute phase of a major depressive episode aims to help the patient reach a remission state and eventually return to their baseline level of functioning. Pharmacotherapy, especially selective serotonin reuptake inhibitors antidepressants, remains the most frequent option for treating depression during the acute phase, while other promising pharmacological options are still competing for the attention of practitioners. Depression-focused psychotherapy is the second most common option for helping patients overcome the acute phase, maintain remission, and prevent relapses. Electroconvulsive therapy is the most effective somatic therapy for depression in some specific situations; meanwhile, other methods have limits, and their specific indications are still being studied. Combining medications, psychotherapy, and somatic therapies remains the most effective way to manage resistant forms of depression.
Collapse
Affiliation(s)
- Rabie Karrouri
- Department of Psychiatry, Moulay Ismaïl Military Hospital, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco
| | - Zakaria Hammani
- Department of Psychiatry, Moulay Ismaïl Military Hospital, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco
| | - Roukaya Benjelloun
- Department of Psychiatry, Faculty of Medicine, Mohammed VI University of Health Sciences, Casablanca 20000, Morocco
| | - Yassine Otheman
- Department of Psychiatry, Moulay Ismaïl Military Hospital, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco
| |
Collapse
|
28
|
Bonaventura J, Lam S, Carlton M, Boehm M, Gomez JL, Solís O, Sánchez-Soto M, Morris PJ, Fredriksson I, Thomas CJ, Sibley DR, Shaham Y, Zarate CA, Michaelides M. Pharmacological and behavioral divergence of ketamine enantiomers: implications for abuse liability. Mol Psychiatry 2021; 26:6704-6722. [PMID: 33859356 PMCID: PMC8517038 DOI: 10.1038/s41380-021-01093-2] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 02/02/2023]
Abstract
Ketamine, a racemic mixture of (S)-ketamine and (R)-ketamine enantiomers, has been used as an anesthetic, analgesic and more recently, as an antidepressant. However, ketamine has known abuse liability (the tendency of a drug to be used in non-medical situations due to its psychoactive effects), which raises concerns for its therapeutic use. (S)-ketamine was recently approved by the United States' FDA for treatment-resistant depression. Recent studies showed that (R)-ketamine has greater efficacy than (S)-ketamine in preclinical models of depression, but its clinical antidepressant efficacy has not been established. The behavioral effects of racemic ketamine have been studied extensively in preclinical models predictive of abuse liability in humans (self-administration and conditioned place preference [CPP]). In contrast, the behavioral effects of each enantiomer in these models are unknown. We show here that in the intravenous drug self-administration model, the gold standard procedure to assess potential abuse liability of drugs in humans, rats self-administered (S)-ketamine but not (R)-ketamine. Subanesthetic, antidepressant-like doses of (S)-ketamine, but not of (R)-ketamine, induced locomotor activity (in an opioid receptor-dependent manner), induced psychomotor sensitization, induced CPP in mice, and selectively increased metabolic activity and dopamine tone in medial prefrontal cortex (mPFC) of rats. Pharmacological screening across thousands of human proteins and at biological targets known to interact with ketamine yielded divergent binding and functional enantiomer profiles, including selective mu and kappa opioid receptor activation by (S)-ketamine in mPFC. Our results demonstrate divergence in the pharmacological, functional, and behavioral effects of ketamine enantiomers, and suggest that racemic ketamine's abuse liability in humans is primarily due to the pharmacological effects of its (S)-enantiomer.
Collapse
Affiliation(s)
- Jordi Bonaventura
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA.
| | - Sherry Lam
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224
| | - Meghan Carlton
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224
| | - Matthew Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224
| | - Juan L. Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224
| | - Oscar Solís
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 21224
| | - Marta Sánchez-Soto
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD, 20892
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850
| | - Ida Fredriksson
- Neurobiology of Relapse Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 212245
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, 20850
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD, 20892
| | - Yavin Shaham
- Neurobiology of Relapse Section, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, 212245
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Intramural Research Program, Bethesda, MD, 20892
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA. .,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
29
|
Heifets BD, Bentzley BS, Williams N, Schatzberg AF. Unraveling the opioid actions of S-ketamine and R-ketamine: comment on Bonaventura et al. Mol Psychiatry 2021; 26:6104-6106. [PMID: 34006965 DOI: 10.1038/s41380-021-01167-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 04/29/2021] [Accepted: 05/06/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Brandon S Bentzley
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Nolan Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan F Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
30
|
Gastaldon C, Raschi E, Kane JM, Barbui C, Schoretsanitis G. Post-Marketing Safety Concerns with Esketamine: A Disproportionality Analysis of Spontaneous Reports Submitted to the FDA Adverse Event Reporting System. PSYCHOTHERAPY AND PSYCHOSOMATICS 2021; 90:41-48. [PMID: 32854103 DOI: 10.1159/000510703] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 07/30/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Esketamine nasal spray received approval for treatment-resistant depression in March 2019. OBJECTIVE Using the FDA Adverse Event Reporting System (FAERS) database (March 2019-March 2020), we analysed esketamine-related adverse events (AEs) to detect and characterize relevant safety signals. METHODS We used the consolidated case/non-case approach to estimate the reporting odds ratio (ROR) and information component (IC) with relevant confidence intervals (95% CI) for esketamine-related AEs with ≥4 counts. Comparisons between serious and non-serious AEs were performed using non-parametric tests. RESULTS The FAERS database contained 962 cases of esketamine-related AEs, with signals detected for several AEs, such as dissociation (ROR = 1,612.64, 95% CI = 1,354.63, 1,919.79; IC = 8.19, 95% CI = 7.96, 8.35), sedation (ROR = 238.46, 95% CI = 202.98, 280.15; IC = 7, 95% CI = 6.75, 7.18), feeling drunk (ROR = 96.17, 95% CI = 61.42, 150.57; IC = 4.84, 95% CI = 4.09, 5.36), suicidal ideation (ROR = 24.03, 95% CI = 18.72, 30.84; IC = 4.31, 95% CI = 3.9, 4.61), and completed suicide (ROR = 5.75, 95% CI = 3.18, 10.41; IC = 2.25, 95% CI = 1.23, 2.94). Signals for suicidal and self-injurious ideation, but not suicide attempt and completed suicide, remained when comparing esketamine to venlafaxine. Females and patients receiving antidepressant polypharmacy, co-medication with mood stabilizers, antipsychotics, benzodiazepines, or somatic medications were more likely to suffer from serious versus non-serious AEs (χ2 = 125.29, p < 0.001, χ2 = 9.08, p = 0.003, χ2 = 8.14, p = 0.004, χ2 = 19.48, p < 0.001, χ2 = 25.62, p < 0.001, and χ2 = 16.79, p < 0.001, respectively). CONCLUSIONS Esketamine may carry a clear potential for serious AEs, which deserves urgent clarification by means of further prospective studies.
Collapse
Affiliation(s)
- Chiara Gastaldon
- WHO Collaborating Centre for Research and Training in Mental Health and Service Evaluation, Department of Neuroscience, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy, .,The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, New York, USA,
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - John M Kane
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, New York, USA.,Department of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,The Feinstein Institute for Medical Research, Center for Psychiatric Neuroscience, Manhasset, New York, USA
| | - Corrado Barbui
- WHO Collaborating Centre for Research and Training in Mental Health and Service Evaluation, Department of Neuroscience, Biomedicine and Movement Sciences, Section of Psychiatry, University of Verona, Verona, Italy
| | - Georgios Schoretsanitis
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, New York, USA
| |
Collapse
|
31
|
Wang XS, Li LC, Zhang X, Gao J. Lipoxin A 4 methyl ester protects PC12 cells from ketamine-induced neurotoxicity via the miR-22/BAG5 pathway. Hum Exp Toxicol 2021; 40:S519-S529. [PMID: 34670429 DOI: 10.1177/09603271211051602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Ketamine is an anesthetic that induces neurotoxicity when administered at high doses. In this work, we explored the protective effects of lipoxin A4 methyl ester (LXA4 ME) against ketamine-induced neurotoxicity and the underlying protective mechanism in pheochromocytoma (PC12) cells. METHODS PC12 cells were treated with 50 μM of ketamine and different LXA4 ME concentrations of LXA4 ME (5-50 nM) for 24 h, and their viability, apoptosis, and oxidative status were assessed. RESULTS Quantitative real-time polymerase chain reaction experiments showed that ketamine downregulated miR-22 expression and upregulated Bcl-2-associated athanogene 5 (BAG5) in PC12 cells in a concentration-dependent manner. LXA4 ME induced the opposite effects, thus attenuating ketamine-induced neurotoxicity. Further in vitro assays showed that miR-22 directly targeted BAG5, thus promoting cell viability by suppressing cell apoptosis and oxidative stress. Under expression miR-22 or upregulation of BAG5 antagonized the effects of LXA4 ME. CONCLUSION LXA4 ME can protect PC12 cells from ketamine-induced neurotoxicity by activating the miR-22/BAG5 signaling pathway. Thus, LXA4 ME can be used as a protective drug against ketamine-induced neural damage.
Collapse
Affiliation(s)
- Xue-Song Wang
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Long-Cheng Li
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xue Zhang
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jin Gao
- Department of Anesthesiology, 74731Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
32
|
Neurocognitive Effects of Ketamine and Esketamine for Treatment-Resistant Major Depressive Disorder: A Systematic Review. Harv Rev Psychiatry 2021; 29:340-350. [PMID: 34366408 DOI: 10.1097/hrp.0000000000000312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
LEARNING OBJECTIVE After participating in this activity, learners should be better able to:• Analyze the effects of ketamine and esketamine on individuals with treatment-resistant depression. INTRODUCTION Cognitive impairment is commonly present in individuals with treatment-resistant depression, especially in attention, memory, and executive functions. These deficits are related to symptom severity, remission rates, and functional impairments during and after the acute phase of the disorder. Ketamine, an N-methyl-D-aspartate antagonist previously used as an anesthetic, brings promising antidepressant results. This study systematically reviews the neurocognitive effects of ketamine and esketamine in patients with treatment-resistant major depressive disorder. METHODS Systematic searches were conducted at Embase, PubMed, and PsycINFO using the terms depression, ketamine, and cognition. Title, abstract, and full-text reading were conducted independently by two of the authors (BSM and CSL). Risk of bias, study design, neuropsychological outcomes, and neuroimaging data were recorded. RESULTS From a total of 997 hits, 14 articles were included. One study reported cognitive impairment after ketamine treatment for processing speed and verbal memory. Five studies reported improvements in processing speed, verbal memory, visual memory, working memory, or cognitive flexibility. The esketamine study suggested no changes to performance. Lower attention, slower processing speed, and higher working memory are reported as predictors of antidepressant response. Brain areas for emotional and reward processing, including the amygdala, insula, and orbitofrontal cortex, show a normalizing tendency after ketamine. CONCLUSIONS Ketamine and esketamine do not seem to exert significant deleterious neurocognitive effects in the short or long term in individuals with treatment-resistant depression. Results suggest neuropsychological functions and brain areas commonly impaired in treatment-resistant depression may especially benefit from subanesthetic ketamine infusions. Key questions that remain unanswered are discussed.
Collapse
|
33
|
Araújo-de-Freitas L, Santos-Lima C, Mendonça-Filho E, Vieira F, França RJAF, Magnavita G, Cardoso TL, Correia-Melo FS, Leal GC, Jesus-Nunes AP, Souza-Marques B, Marback R, Teles M, Echegaray MV, Beanes G, Guerreiro-Costa LNF, Mello RP, Rabanea T, Lucchese AC, Abreu N, Lacerda ALT, Quarantini LC. Neurocognitive aspects of ketamine and esketamine on subjects with treatment-resistant depression: A comparative, randomized and double-blind study. Psychiatry Res 2021; 303:114058. [PMID: 34153630 DOI: 10.1016/j.psychres.2021.114058] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 06/05/2021] [Indexed: 01/28/2023]
Abstract
The objective of this study is to evaluate cognition in patients using either ketamine or esketamine to treat TRD. We also evaluate if both ketamine and esketamine as one group influence cognition in patients with TRD. Fifty-four patients with TRD were infused with either ketamine or esketamine and were assessed at three time points: baseline, 24 h, and 7 days after infusion. We applied neuropsychological tests to evaluate executive functions, processing speed, short term memory, and auditory-verbal episodic memory. There is no cognitive difference between ketamine and esketamine, with the exception of one variable. When considered as one group, ketamine and esketamine do not impair cognition; on the contrary, they improve some neuropsychological functions such as visuospatial short-term memory, executive functions, processing speed, and several measures related to episodic verbal memory. Ketamine and esketamine do not present differing cognitive effects when used in antidepressant doses to treat TRD. Furthermore, they rapidly improve many cognitive aspects of patients with TRD at 24 h after the infusion and maintain these effects for at least 7 days.
Collapse
Affiliation(s)
- Lucas Araújo-de-Freitas
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil; Departamento de Neurociências e Saúde Mental, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Cassio Santos-Lima
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil; Laboratório de Pesquisa em Neuropsicologia Clínica e Cognitiva, UFBA, Salvador, Brazil; Programa de Pós-graduação em Psicologia, Instituto de Psicologia, UFBA, Salvador, Brazil
| | - Euclides Mendonça-Filho
- Programa de Pós-graduação em Psicologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Department of Psychiatry, McGill University, Montreal, Canada
| | - Flávia Vieira
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil
| | - Ricardo J A F França
- Laboratório de Pesquisa em Neuropsicologia Clínica e Cognitiva, UFBA, Salvador, Brazil; Programa de Pós-graduação em Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Guilherme Magnavita
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Tanise L Cardoso
- Laboratório de Pesquisa em Neuropsicologia Clínica e Cognitiva, UFBA, Salvador, Brazil; Programa de Pós-graduação em Psicologia, Instituto de Psicologia, UFBA, Salvador, Brazil
| | - Fernanda S Correia-Melo
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Gustavo C Leal
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Ana Paula Jesus-Nunes
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil
| | - Breno Souza-Marques
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil
| | - Roberta Marback
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Manuela Teles
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil
| | - Mariana Vf Echegaray
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Graziele Beanes
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil
| | - Lívia N F Guerreiro-Costa
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil
| | - Rodrigo P Mello
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil
| | - Thaís Rabanea
- Laboratório Interdisciplinar de Neurociências Clínicas, Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Psiquiatria, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Cecília Lucchese
- Programa de Pós-graduação em Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil; Laboratório Interdisciplinar de Neurociências Clínicas, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Neander Abreu
- Laboratório de Pesquisa em Neuropsicologia Clínica e Cognitiva, UFBA, Salvador, Brazil; Programa de Pós-graduação em Psicologia, Instituto de Psicologia, UFBA, Salvador, Brazil
| | - Acioly L T Lacerda
- Programa de Pós-graduação em Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil; Laboratório Interdisciplinar de Neurociências Clínicas, Universidade Federal de São Paulo, São Paulo, Brazil; Departamento de Psiquiatria, Universidade Federal de São Paulo, São Paulo, Brazil; Instituto Sinapse de Neurociências Clínicas, Campinas, Brazil
| | - Lucas C Quarantini
- Laboratório de Neuropsicofarmacologia, Serviço de Psiquiatria, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador, Brazil; Programa de Pós-graduação em Medicina e Saúde, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil; Departamento de Neurociências e Saúde Mental, Faculdade de Medicina da Bahia, UFBA, Salvador, Brazil.
| |
Collapse
|
34
|
Van Amsterdam J, Van Den Brink W. Harm related to recreational ketamine use and its relevance for the clinical use of ketamine. A systematic review and comparison study. Expert Opin Drug Saf 2021; 21:83-94. [PMID: 34176409 DOI: 10.1080/14740338.2021.1949454] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUNDS Ketamine is a dissociative anesthetic that is currently considered for several new indications. AIM To deduce the safety of long-term ketamine treatment using the harm of heavy recreational (non-medical) ketamine use as a proxy for maximal possible harm of ketamine treatment. METHODS Systematic literature review according to PRISMA guidelines to identify controlled studies on ketamine-related harm in heavy recreational ketamine users. Results were compared with serious adverse events (SAEs) in patients treated with ketamine according to three systematic reviews considering dosing regimen and cumulative dose. RESULTS The systematic search yielded 25 studies. Heavy recreational ketamine use can escalate to ketamine dependency and was often dose-dependently associated with other SAEs, including cognitive and mental disorders, and gastrointestinal and urinary tract symptoms, which disappeared upon marked reduction of ketamine use. Heavy ketamine users have a much higher cumulative exposure to ketamine than ketamine treated patients (>90 times), which may explain why SAEs in the clinical context are mostly mild and reversible and why ketamine dependence was not reported in these patients. CONCLUSION Treatment of patients with ketamine is not associated with ketamine dependency or SAEs. However, caution is needed since data on long-term clinical ketamine use with a long-term follow-up is lacking.
Collapse
Affiliation(s)
- Jan Van Amsterdam
- Department of Psychiatry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Wim Van Den Brink
- Department of Psychiatry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Cosci F, Chouinard G. Acute and Persistent Withdrawal Syndromes Following Discontinuation of Psychotropic Medications. PSYCHOTHERAPY AND PSYCHOSOMATICS 2021; 89:283-306. [PMID: 32259826 DOI: 10.1159/000506868] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/27/2020] [Indexed: 11/19/2022]
Abstract
Studies on psychotropic medications decrease, discontinuation, or switch have uncovered withdrawal syndromes. The present overview aimed at analyzing the literature to illustrate withdrawal after decrease, discontinuation, or switch of psychotropic medications based on the drug class (i.e., benzodiazepines, nonbenzodiazepine benzodiazepine receptor agonists, antidepressants, ketamine, antipsychotics, lithium, mood stabilizers) according to the diagnostic criteria of Chouinard and Chouinard [Psychother Psychosom. 2015;84(2):63-71], which encompass new withdrawal symptoms, rebound symptoms, and persistent post-withdrawal disorders. All these drugs may induce withdrawal syndromes and rebound upon discontinuation, even with slow tapering. However, only selective serotonin reuptake inhibitors, serotonin noradrenaline reuptake inhibitors, and antipsychotics were consistently also associated with persistent post-withdrawal disorders and potential high severity of symptoms, including alterations of clinical course, whereas the distress associated with benzodiazepines discontinuation appears to be short-lived. As a result, the common belief that benzodiazepines should be substituted by medications that cause less dependence such as antidepressants and antipsychotics runs counter the available literature. Ketamine, and probably its derivatives, may be classified as at high risk for dependence and addiction. Because of the lag phase that has taken place between the introduction of a drug into the market and the description of withdrawal symptoms, caution is needed with the use of newer antidepressants and antipsychotics. Within medication classes, alprazolam, lorazepam, triazolam, paroxetine, venlafaxine, fluphenazine, perphenazine, clozapine, and quetiapine are more likely to induce withdrawal. The likelihood of withdrawal manifestations that may be severe and persistent should thus be taken into account in clinical practice and also in children and adolescents.
Collapse
Affiliation(s)
- Fiammetta Cosci
- Department of Health Sciences, University of Florence, Florence, Italy, .,Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,
| | - Guy Chouinard
- Clinical Pharmacology and Toxicology Program, McGill University and Mental Health Institute of Montreal Fernand Seguin Research Centre, University of Montreal, Montreal, Québec, Canada
| |
Collapse
|
36
|
Targeting the dysfunction of glutamate receptors for the development of novel antidepressants. Pharmacol Ther 2021; 226:107875. [PMID: 33901503 DOI: 10.1016/j.pharmthera.2021.107875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 12/19/2022]
Abstract
Increasing evidence indicates that dysfunction of glutamate receptors is involved in the pathophysiology of major depressive disorder (MDD). Although accumulating efforts have been made to elucidate the applications and mechanisms underlying antidepressant-like effects of ketamine, a non-selective antagonist of N-methyl-d-aspartate receptor (NMDAR), the role of specific glutamate receptor subunit in regulating depression is not completely clear. The current review aims to discuss the relationships between glutamate receptor subunits and depressive-like behaviors. Research literatures were searched from inception to July 2020. We summarized the alterations of glutamate receptor subunits in patients with MDD and animal models of depression. Animal behaviors in response to dysfunction of glutamate receptor subunits were also surveyed. To fully understand mechanisms underlying antidepressant-like effects of modulators targeting glutamate receptors, we discussed effects of each glutamate receptor subunit on serotonin system, synaptic plasticity, neurogenesis and neuroinflammation. Finally, we collected most recent clinical applications of glutamate receptor modulators and pointed out the limitations of these candidates in the treatment of MDD.
Collapse
|
37
|
Kaur U, Pathak BK, Singh A, Chakrabarti SS. Esketamine: a glimmer of hope in treatment-resistant depression. Eur Arch Psychiatry Clin Neurosci 2021; 271:417-429. [PMID: 31745646 DOI: 10.1007/s00406-019-01084-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022]
Abstract
The motive of this article is to review the pharmacological and clinical aspects of esketamine (ESK), an NMDA-receptor antagonist approved recently by the FDA for treatment-resistant depression (TRD). PubMed/Medline database was searched using keywords 'esketamine' and 'depression', 'S-ketamine' and 'depression', and 'NMDA antagonist' and 'depression'. Individual trials were searched from ClinicalTrials.gov. We included English-language articles evaluating pharmacokinetics and pharmacodynamics of intranasal (IN) esketamine, along with clinical trial data related to its efficacy and safety in patients diagnosed with TRD. Compared to placebo, IN esketamine causes significant and rapid improvement in depression. Dizziness, vertigo, headache, increase in blood pressure are some of its common adverse effects. With the growing number of patients of TRD, additional effective and safe treatment is the need of the hour. Esketamine appears to be an effective therapy when combined with oral antidepressants in patients with TRD. It is of special value due to the rapid onset of its action. Long-term clinical studies are, however, needed to ascertain its safety profile.
Collapse
Affiliation(s)
- Upinder Kaur
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India
| | - Bhairav Kumar Pathak
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India
| | - Amit Singh
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India
| | - Sankha Shubhra Chakrabarti
- Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP, India.
| |
Collapse
|
38
|
Esses G, Deiner S, Ko F, Khelemsky Y. Chronic Post-Surgical Pain in the Frail Older Adult. Drugs Aging 2020; 37:321-329. [PMID: 32297246 DOI: 10.1007/s40266-020-00761-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Older adults are the fastest growing segment of the population and surgical procedures in this group increase each year. Chronic post-surgical pain is an important consideration in the older adult as it affects recovery, physical functioning, and overall quality of life. It is increasingly recognized as a public health issue but there is a need to improve our understanding of the disease process as well as the appropriate treatment and prevention. Frailty, delirium, and cognition influence post-operative outcomes in older adults and have been implicated in the development of chronic post-surgical pain. Further research must be conducted to fully understand the role they play in the occurrence of chronic post-surgical pain in the older adult. Additionally, careful attention must be given to the physiologic, cognitive, and comorbidity differences between the older adult and the general population. This is critical for elucidating the proper chronic post-surgical pain treatment and prevention strategies to ensure that the older adult undergoing surgical intervention will have an appropriate and desirable post-operative outcome.
Collapse
Affiliation(s)
- Gary Esses
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, Box 1010, New York, NY, USA.
| | - Stacie Deiner
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, Box 1010, New York, NY, USA
| | - Fred Ko
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yury Khelemsky
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, Box 1010, New York, NY, USA
| |
Collapse
|
39
|
Wang P, Yang Z, Shan S, Cao Z, Wang Z. Analgesic effect of perioperative ketamine for total hip arthroplasties and total knee arthroplasties: A PRISMA-compliant meta-analysis. Medicine (Baltimore) 2020; 99:e22809. [PMID: 33080757 PMCID: PMC7571980 DOI: 10.1097/md.0000000000022809] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Total hip arthroplasties (THA) and total knee arthroplasties (TKA) are always associated with a frequent incidence of postoperative pain. Effective pain management after surgery is quite essential for surgeons and patients. The purpose of the present meta-analysis is to evaluate the analgesic effect of perioperative ketamine after THA and TKA. METHODS Seven online databases, Embase, Cochrane Library, Pubmed, Web of Science, China National Knowledge Infrastructure (CNKI), China Biomedical Literature Database (CBM), and Wanfang Data were searched for the related randomized controlled trials (RCT) by August 15, 2019. The qualities of the included studies were assessed based on the Cochrane Handbook for Systematic Reviews of Interventions 5.0. The visual analog scale (VAS), morphine equivalent consumption, and the side effects were used to evaluate the postoperative analgesic effect of ketamine by meta-analysis, which was performed by Review Manager version 5.3 software. RESULTS The VAS scores at 6 hours, 12 hours, 24 hours, and 48 hours after surgery were statistically lower in the ketamine group. The morphine equivalent consumptions in 24 hours and 48 hours after surgery were also significantly lower in the ketamine group. For the side effects, no statistical differences in odds ratio (OR) of sedation, dizziness, hallucination, sweating, pruritus, urinary retention, constipation, version trouble, nightmares, and delirium were observed between the ketamine group and the control group. But postoperative nausea and vomiting (PONV) showed lower OR in the ketamine group. CONCLUSION The present meta-analysis demonstrated perioperative ketamine could be used as a safe and effective analgesic agent for THA and TKA.
Collapse
Affiliation(s)
| | - Zhong Yang
- Department of Orthopedics, The Fifth Central Hospital of Tianjian, Tianjin
| | | | - Zhipeng Cao
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province
| | - Zhilin Wang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, PR China
| |
Collapse
|
40
|
Xia SH, Hu SW, Ge DG, Liu D, Wang D, Zhang S, Zhang Q, Yuan L, Li YQ, Yang JX, Wu P, Zhang H, Han MH, Ding HL, Cao JL. Chronic Pain Impairs Memory Formation via Disruption of Neurogenesis Mediated by Mesohippocampal Brain-Derived Neurotrophic Factor Signaling. Biol Psychiatry 2020; 88:597-610. [PMID: 32307038 DOI: 10.1016/j.biopsych.2020.02.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/30/2020] [Accepted: 02/14/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Chronic pain patients often complain of their poor memory. The mechanisms underlying chronic pain-related memory impairment remain elusive, and there are few clinical therapeutic strategies available for this condition. METHODS In a neuropathic pain model induced by chronic constrictive injury of the sciatic nerve in male mice, we used circuit-specific electrophysiological recording, combined with chemogenetic, molecular, and pharmacologic methods, to examine the circuit and molecular mechanisms underlying chronic pain-related memory impairment. RESULTS Our current results show that chronic neuropathic pain impaired the acquisition of spatial memory and, meanwhile, reduced adult neurogenesis in the dentate gyrus. Experimentally reducing dentate gyrus neurogenesis mimicked this pain-induced effect on spatial memory formation in naïve mice. Furthermore, pain-associated impairments of both hippocampal neurogenesis and memory formation were rescued or mimicked by chemogenetic activation or deactivation, respectively, of the ventral tegmental area dopaminergic projection, through which ventral tegmental area-released brain-derived neurotrophic factor was required. Importantly, we found that chronic, but not acute, systematic administration of subanesthetic doses of ketamine, while without relieving pain, ameliorated chronic pain-related impairment of spatial memory formation, potentially by rescuing brain-derived neurotrophic factor-mediated dentate gyrus neurogenesis. CONCLUSIONS These findings provide a novel, circuit-based mechanistic link between chronic pain and memory formation deficit, and potential new therapeutic options for chronic pain-related learning deficit and memory impairment.
Collapse
Affiliation(s)
- Sun-Hui Xia
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - De-Gao Ge
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Di Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Di Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Song Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Qi Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Ling Yuan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Yan-Qiang Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Peng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Ming-Hu Han
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York; Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China; Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
41
|
Farmer CA, Gilbert JR, Moaddel R, George J, Adeojo L, Lovett J, Nugent AC, Kadriu B, Yuan P, Gould TD, Park LT, Zarate CA. Ketamine metabolites, clinical response, and gamma power in a randomized, placebo-controlled, crossover trial for treatment-resistant major depression. Neuropsychopharmacology 2020; 45:1398-1404. [PMID: 32252062 PMCID: PMC7297997 DOI: 10.1038/s41386-020-0663-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 12/27/2022]
Abstract
A single, subanesthetic dose of (R,S)-ketamine (ketamine) exerts rapid and robust antidepressant effects. Several groups previously reported that (2S,6S;2R,6R)-hydroxynorketamine (HNK) had antidepressant effects in rodents, and that (2R,6R)-HNK increased cortical electroencephalographic gamma power. This exploratory study examined the relationship between ketamine metabolites, clinical response, psychotomimetic symptoms, and gamma power changes in 34 individuals (ages 18-65) with treatment-resistant depression (TRD) who received a single ketamine infusion (0.5 mg/kg) over 40 min. Plasma concentrations of ketamine, norketamine, and HNKs were measured at 40, 80, 120, and 230 min and at 1, 2, and 3 days post-infusion. Linear mixed models evaluated ketamine metabolites as mediators of antidepressant and psychotomimetic effects and their relationship to resting-state whole-brain magnetoencephalography (MEG) gamma power 6-9 h post-infusion. Three salient findings emerged. First, ketamine concentration positively predicted distal antidepressant response at Day 11 post-infusion, and an inverse relationship was observed between (2S,6S;2R,6R)-HNK concentration and antidepressant response at 3 and 7 days post-infusion. Norketamine concentration was not associated with antidepressant response. Second, ketamine, norketamine, and (2S,6S;2R,6R)-HNK concentrations at 40 min were positively associated with contemporaneous psychotomimetic symptoms; post-hoc analysis revealed that ketamine was the predominant contributor. Third, increased (2S,6S;2R,6R)-HNK maximum observed concentration (Cmax) was associated with increased MEG gamma power. While contrary to preclinical observations and our a priori hypotheses, these exploratory results replicate those of a recently published study documenting a relationship between higher (2S,6S;2R,6R)-HNK concentrations and weaker antidepressant response in humans and provide further rationale for studying gamma power changes as potential biomarkers of antidepressant response.
Collapse
Affiliation(s)
- Cristan A Farmer
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Jessica R Gilbert
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ruin Moaddel
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jomy George
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Lilian Adeojo
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Jacqueline Lovett
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Allison C Nugent
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Magnetoencephalography Core Facility, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Peixiong Yuan
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Departments of Psychiatry, Pharmacology, and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Lawrence T Park
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
42
|
Lavender E, Hirasawa-Fujita M, Domino EF. Ketamine's dose related multiple mechanisms of actions: Dissociative anesthetic to rapid antidepressant. Behav Brain Res 2020; 390:112631. [PMID: 32437885 DOI: 10.1016/j.bbr.2020.112631] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 03/19/2020] [Accepted: 03/29/2020] [Indexed: 01/15/2023]
Abstract
Ketamine induces safe and effective anesthesia and displays unusual cataleptic properties that gave rise to the term dissociative anesthesia. Since 1970, clinicians only utilized the drug as an anesthetic or analgesic for decades, but ketamine was found to have rapid acting antidepressant effects in 1990s. Accumulated evidence exhibits NMDAR antagonism may not be the only mechanism of ketamine. The contributions of AMPA receptor, mTor signal pathway, monoaminergic system, sigma-1 receptor, cholinergic, opioid and cannabinoid systems, as well as voltage-gated calcium channels and hyperpolarization cyclic nucleotide gated channels are discussed for the antidepressant effects. Also the effects of ketamine's enantiomers and metabolites are reviewed. Furthermore ketamine's anesthetic and analgesic mechanisms are briefly revisited. Overall, pharmacology of ketamine, its enantiomers and metabolites is very unique. Insight into multiple mechanisms of action will provide further development and desirable clinical effects of ketamine.
Collapse
Affiliation(s)
- Eli Lavender
- University of Michigan Medical School, Department of Pharmacology, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Mika Hirasawa-Fujita
- University of Michigan Medical School, Department of Pharmacology, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA
| | - Edward F Domino
- University of Michigan Medical School, Department of Pharmacology, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA.
| |
Collapse
|
43
|
Abstract
Major depressive disorder (MDD) is a debilitating illness with significant morbidity and mortality, leading to attempted and completed suicides. It affects interpersonal relationships and also contributes to decreased productivity, causing financial burden to individuals and society. Patients often fail to respond to various antidepressant medication trials resulting in treatment-resistant depression (TRD). Current antidepressant medications work by modulating the monoaminergic systems and takes several weeks to establish a clinical response. Ketamine has been used extensively as an anesthetic agent since the 1970s, and more recent research has shown its rapid and robust effectiveness in TRD the subject of this review. Ketamine is a racemic mixture comprised of two enantiomers (R)-ketamine and (S)-ketamine and acts as an NMDA receptor antagonist. Most research studies have explored its antidepressant and antisuicidal effects by using it as an intravenous infusion or via the intranasal route due to increased bioavailability. Recently an intranasal esketamine spray was approved by the United States Food and Drug Administration (FDA) for TRD as an adjunct to standard antidepressant treatment in a supervised setting. Regarding its safety profile, multiple research studies have established the short-term safety and efficacy of ketamine in TRD. The cardiorespiratory and neuropsychiatric adverse events observed in these studies were mostly transient. However, ketamine is a scheduled agent with abuse potential, making its long-term use challenging and mandating further research.
Collapse
|
44
|
Pérez-Esparza R, Kobayashi-Romero LF, García Mendoza AM, Lamas-Aguilar RM, Vargas Sosa M, Encarnación-Martínez M, González-Manríquez LA, Eternod-Rodríguez SA, Maltos-Gómez F, Vargas-Valencia KM, Fonseca Pérez-Amador A. Ketamina, un nuevo agente terapéutico para la depresión. REVISTA DE LA FACULTAD DE MEDICINA 2020. [DOI: 10.22201/fm.24484865e.2020.63.1.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder affects about one in every 10 people in Mexico and is one of the first 5 causes of disability worldwide. Current treatment options are limited and only act upon some factors associated in its physiopathology. Moreover, the effects on depression are not immediate, which is a great limitation in obtaining a benefit over disability caused by this disorder and impedes a rapid action in the scenario of suicidality. Recently, ketamine (an anesthetic) has shown to have antidepressant properties by acting in the glutamate neurotransmission system (while no other current treatment acts on this level). It offers benefits in depressive symptoms in a matter of hours and has proven to be useful in patients that do not benefit from current therapeutic options. Recently, it has been approved for the treatment of depression. However, there are still many questions about its antidepressant mechanisms of action, safety, side effects, among others. Key words: Depression; antidepressants; ketamine.
Collapse
Affiliation(s)
- Rodrigo Pérez-Esparza
- Secretarìa de Salud (Ssa), Instituto Nacional de Neurología y Neurocirugía. Laboratorio de Investigación en Adicciones. Ciudad de México, México.; Secretarìa de Salud (Ssa), Instituto Nacional de Neurología y Neurocirugía. Departamento de Neuropsiquiatría. Ciudad de México, México
| | - Luis Fabián Kobayashi-Romero
- Secretarìa de Salud (Ssa), Instituto Nacional de Neurología y Neurocirugía. Laboratorio de Investigación en Adicciones. Ciudad de México, México
| | - Ana María García Mendoza
- Secretarìa de Salud (Ssa), Instituto Nacional de Neurología y Neurocirugía. Laboratorio de Investigación en Adicciones. Ciudad de México, México; Universidad Nacional Autónoma de México (UNAM), Facultad de Medicina, Ciudad de México, México
| | - Reyna Minerva Lamas-Aguilar
- Secretarìa de Salud (Ssa), Instituto Nacional de Neurología y Neurocirugía. Departamento de Neuropsiquiatría. Ciudad de México, México
| | - Melissa Vargas Sosa
- Universidad Nacional Autónoma de México (UNAM), Facultad de Medicina. Ciudad de México, México
| | | | | | | | - Fernanda Maltos-Gómez
- Universidad Nacional Autónoma de México (UNAM), Facultad de Medicina. Ciudad de México, México
| | | | - Alan Fonseca Pérez-Amador
- Secretarìa de Salud (Ssa), Instituto Nacional de Neurología y Neurocirugía. Laboratorio de Investigación en Adicciones. Ciudad de México, México
| |
Collapse
|
45
|
Pompili M. Intranasal Esketamine and Current Suicidal Ideation With Intent in Major Depression Disorder: Beat the Clock, Save a Life, Start a Strategy. Front Psychiatry 2020; 11:325. [PMID: 32425826 PMCID: PMC7203410 DOI: 10.3389/fpsyt.2020.00325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/01/2020] [Indexed: 01/08/2023] Open
Affiliation(s)
- Maurizio Pompili
- Department of Neuroscience, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
46
|
|
47
|
Tan Y, Fujita Y, Qu Y, Chang L, Pu Y, Wang S, Wang X, Hashimoto K. Phencyclidine-induced cognitive deficits in mice are ameliorated by subsequent repeated intermittent administration of (R)-ketamine, but not (S)-ketamine: Role of BDNF-TrkB signaling. Pharmacol Biochem Behav 2019; 188:172839. [PMID: 31866390 DOI: 10.1016/j.pbb.2019.172839] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 01/25/2023]
Abstract
The N-methyl-d-aspartate receptor (NMDAR) antagonists including phencyclidine (PCP) and ketamine produce cognitive deficits in rodents and humans. We previously reported that (R)-ketamine produced the beneficial effects compared to (S)-ketamine in several animal models including depression. Here we compared the effects of two enantiomers of ketamine on cognitive deficits in mice after repeated administration of PCP. PCP (10 mg/kg/day for 10 days)-induced cognitive deficits were ameliorated by subsequent repeated intermittent administration of (R)-ketamine (10 mg/kg/day, twice weekly for 2-weeks), but not (S)-ketamine. Western blot analysis showed decreased levels of brain-derived neurotrophic factor (BDNF) and decreased ratio of phosphorylated-TrkB (p-TrkB) to TrkB in the prefrontal cortex (PFC) and hippocampus of PCP-treated mice. Furthermore, PCP-induced reduction of BDNF and p-TrkB/TrkB ratio in the PFC and hippocampus of PCP-treated mice was ameliorated by subsequent intermittent administration of (R)-ketamine. Interestingly, the beneficial effects of (R)-ketamine were blocked by pretreatment with TrkB inhibitor ANA-12. These findings suggest that (R)-ketamine could ameliorate PCP-induced cognitive deficits via activation of BDNF-TrkB signaling in the brain. Therefore, (R)-ketamine could be a potential therapeutic drug for cognitive impairment in patients with schizophrenia.
Collapse
Affiliation(s)
- Yunfei Tan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Youge Qu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yaoyu Pu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Siming Wang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Xingming Wang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan.
| |
Collapse
|
48
|
Abstract
In March 2019, the US Food and Drug Administration (FDA) approved a nasal spray formulation of esketamine for the treatment of resistant depression in adults. Esketamine is the S-enantiomer of ketamine, an FDA-approved anaesthetic, known to cause dissociation and, occasionally, hallucinations. While ketamine has not been approved for depression in the USA or in any other country, it has been used off-label in cases of severe depression. This commentary critically reviewed the evidence on esketamine submitted to the FDA, aiming to draw implications for clinical practice, research and regulatory science.
Collapse
|
49
|
Huang N, Wang Y, Zhan G, Yu F, Li S, Hua D, Jiang R, Li S, Wu Y, Yang L, Zhu B, Hua F, Luo A, Yang C. Contribution of skeletal muscular glycine to rapid antidepressant effects of ketamine in an inflammation-induced mouse model of depression. Psychopharmacology (Berl) 2019; 236:3513-3523. [PMID: 31321459 DOI: 10.1007/s00213-019-05319-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/30/2019] [Indexed: 01/09/2023]
Abstract
RATIONALE Basic and clinical studies have reported rapid and long-lasting antidepressant effects of ketamine. Although previous studies have proposed several mechanisms underlying the antidepressant effects of ketamine, these mechanisms have not been completely elucidated. OBJECTIVES The present study evaluated the effects of systemically administered ketamine treatment in a lipopolysaccharide (LPS)-induced mouse model of depression. METHODS Non-targeted metabolomics, western blotting, and behavioral tests (locomotion, tail suspension, and forced swimming tests) were performed. RESULT Ketamine significantly attenuated the abnormally increased immobility time in a lipopolysaccharide (LPS)-induced mouse model of depression. Aminomalonic acid, glutaraldehyde, glycine, histidine, N-methyl-L-glutamic acid, and ribose levels in skeletal muscle were altered following ketamine administration. Furthermore, ketamine significantly decreased the LPS-induced increase in glycine receptor A1 (GlyA1) levels. However, the glycine receptor antagonist strychnine did not elicit any pharmacological effects on ketamine-induced alterations in behaviors or muscular GlyA1 levels. Exogenous glycine and L-serine significantly improved depression-like symptoms in LPS-induced mice. CONCLUSIONS Our findings suggest that skeletal muscular glycine contributes to the antidepressant effects of ketamine in inflammation. Effective strategies for improving skeletal muscular glycine levels may be a novel approach to depression treatment.
Collapse
Affiliation(s)
- Niannian Huang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Yue Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Fan Yu
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Shan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Dongyu Hua
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Riyue Jiang
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Yeshun Wu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Chun Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
50
|
Antidepressant effects of ketamine on depression-related phenotypes and dopamine dysfunction in rodent models of stress. Behav Brain Res 2019; 379:112367. [PMID: 31739001 DOI: 10.1016/j.bbr.2019.112367] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
Depression, the most prevalent psychiatric disorder, is characterized by increased negative affect (i.e. depressed mood) and reduced positive affect (i.e. anhedonia). Stress is a risk factor for depression in humans, and animal models of chronic stress are typically used to study neurobehavioral alterations relevant to depression. Common behavioral outcomes in rodent models of chronic stress include anhedonia, social dysfunction and behavioral despair. For example, chronically stressed rodents exhibit reduced reward preference, as measured by a loss of preference for sucrose solutions and time spent interacting with a novel conspecific, while also exhibiting less time struggling against inescapable stressors (e.g. forced swim, tail suspension). In both humans and rodents, anhedonia is associated with dysfunction of the dopamine (DA) system. Unlike traditional antidepressants, which are limited by inadequate efficacy and delayed therapeutic response, acute ketamine administration rapidly alleviates depressive symptoms in humans and reverses stress-induced changes in animal models. These effects are partially mediated via actions on the DA system. This review summarizes the clinical effects of ketamine, the neurobiological underpinnings of depression with a focus on DA dysfunction, as well as antidepressant effects of ketamine on depression-related endophenotypes (i.e. anhedonia, despair) and ventral tegmental area (VTA) activity in rodent models of repeated stress. Moreover, we discuss evidence regarding sex differences in ketamine's antidepressant effects, wherein females appear to be more sensitive to lower dose ketamine, as well as novel findings suggesting that ketamine has prophylactic effects with regard to protection against the neurobehavioral impact of future stressors.
Collapse
|