1
|
Nagamine T. Merits and demerits of administering esketamine in preventing postpartum depression following cesarean section. World J Clin Cases 2024; 12:6883-6886. [PMID: 39726928 PMCID: PMC11531973 DOI: 10.12998/wjcc.v12.i36.6883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/01/2024] [Accepted: 09/10/2024] [Indexed: 10/31/2024] Open
Abstract
Emergency cesarean section is associated with the development of postpartum depression. Esketamine has been demonstrated to have a rapid onset of antidepressant effects. Randomized controlled trials and meta-analyses have demonstrated the efficacy of esketamine in preventing postpartum depression after cessarean section. However, the data included in these analyses were derived from elective cesarean sections and differed in the dose and timing of esketamine administration. Esketamine is a dissociative anesthetic with a dose-dependent risk of inducing psychotic symptoms, including hallucinations. In the setting of cesarean section, esketamine should be administered with caution and only if the potential benefits outweigh the risks.
Collapse
Affiliation(s)
- Takahiko Nagamine
- Department of Psychiatric Internal Medicine, Sunlight Brain Research Center, Hofu 7470066, Yamaguchi, Japan
| |
Collapse
|
2
|
Yang C, Ali T, Li A, Gao R, Yu X, Li S, Li T. Ketamine reverses chronic corticosterone-induced behavioral deficits and hippocampal synaptic dysfunction by regulating eIF4E/BDNF signaling. Neuropharmacology 2024; 261:110156. [PMID: 39326783 DOI: 10.1016/j.neuropharm.2024.110156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
Major depressive disorder (MDD) is a debilitating illness with a high global burden. While Ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, offers rapid-acting antidepressant effects, its mechanism remains incompletely understood. Recent research suggests that dysregulation of mRNA translation via the Eukaryotic initiation factor 4E (eIF4E) pathway might contribute to depression pathophysiology. This study investigates whether Ketamine modulates eIF4E signaling in the hippocampus during its antidepressant action. Herein, adult male mice were exposed to Corticosterone, a well-established model for anxiety and depression, followed by behavioral testing and biochemical analysis. Corticosterone induced depression-like symptoms and disrupted synaptic function, including reduced TrkB/BDNF and eIF4E/MNK1/p-eIF2α/ubiquitin signaling. Ketamine treatment reversed these deficits. Notably, the eIF4E/MNK1 signaling inhibitor, eFT508, blocked Ketamine's antidepressant effect, leading to a return of depression-like phenotype and impaired synaptic signaling. Importantly, these effects were reversed by 7,8-DHF, a BDNF/TrkB signaling agonist. Mice treated with Corticosterone, Ketamine, and eFT508 and subsequently exposed to 7,8-DHF displayed normalized depression-like behaviors and restored synaptic signaling, including increased eIF4E phosphorylation and MNK1 expression. Besides, 7,8-DHF treatment enhanced p-eIF2α levels compared to the eFT508-treated group. These findings suggest that Ketamine exerts its antidepressant action through the regulation of the eIF4E/BDNF signaling pathway in the hippocampus. This study provides novel insights into the molecular mechanisms underlying Ketamine's therapeutic effects and highlights the potential of targeting this pathway for future MDD treatment strategies.
Collapse
Affiliation(s)
- Canyu Yang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China; State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Tahir Ali
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China; Shenzhen Bay Laboratory, Shenzhen 518055, China.
| | - Axiang Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China; State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Ruyan Gao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Xiaoming Yu
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, People's Republic of China.
| | - Shupeng Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China; Shenzhen Bay Laboratory, Shenzhen 518055, China; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| | - Tao Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, China; NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University. Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
3
|
Gao Z, Winhusen TJ, Gorenflo MP, Dorney I, Ghitza UE, Kaelber DC, Xu R. Artificial intelligence-based drug repurposing with electronic health record clinical corroboration: A case for ketamine as a potential treatment for amphetamine-type stimulant use disorder. Addiction 2024. [PMID: 39552271 DOI: 10.1111/add.16715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND AND AIMS Amphetamine-type stimulants are the second-most used illicit drugs globally, yet there are no US Food and Drug Administration (FDA)-approved treatments for amphetamine-type stimulant use disorders (ATSUD). The aim of this study was to utilize a drug discovery framework that integrates artificial intelligence (AI)-based drug prediction, clinical corroboration and mechanism of action analysis to identify FDA-approved drugs that can be repurposed for treating ATSUD. DESIGN AND SETTING An AI-based knowledge graph model was first utilized to prioritize FDA-approved drugs in their potential efficacy for treating ATSUD. Among the top 10 ranked candidate drugs, ketamine represented a novel candidate with few studies examining its effects on ATSUD. We therefore conducted a retrospective cohort study to assess the association between ketamine and ATSUD remission using US electronic health record (EHR) data. Finally, we analyzed the potential mechanisms of action of ketamine in the context of ATSUD. PARTICIPANTS AND MEASUREMENTS ATSUD patients who received anesthesia (n = 3663) or were diagnosed with depression (n = 4328) between January 2019 and June 2022. The outcome measure was the diagnosis of ATSUD remission within one year of the drug prescription. FINDINGS Ketamine for anesthesia in ATSUD patients was associated with greater ATSUD remission compared with other anesthetics: hazard ratio (HR) = 1.58, 95% confidence interval (CI) = 1.15-2.17. Similar results were found for ATSUD patients with depression when comparing ketamine with antidepressants and bupropion/mirtazapine with HRs of 1.51 (95% CI = 1.14-2.01) and 1.68 (95% CI = 1.18-2.38), respectively. Functional analyses demonstrated that ketamine targets several ATSUD-associated pathways including neuroactive ligand-receptor interaction and amphetamine addiction. CONCLUSIONS There appears to be an association between clinician-prescribed ketamine and higher remission rates in patients with amphetamine-type stimulant use disorders.
Collapse
Affiliation(s)
- Zhenxiang Gao
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - T John Winhusen
- Center for Addiction Research, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Maria P Gorenflo
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ian Dorney
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Udi E Ghitza
- Center for the Clinical Trials Network (CCTN), National Institute on Drug Abuse (NIDA), National Institutes of Health (NIH), Bethesda, MD, USA
| | - David C Kaelber
- Center for Clinical Informatics Research and Education, The Metro Health System, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
4
|
Martire G, Sipple D, Baron D, Gold MS, Lewandowski KU, Dennen CA, Sharafshah A, Elman I, Thanos PK, Modestino EJ, Badgaiyan RD, Pinhasov A, Bowirrat A, Makale M, Roy AK, Sunder K, Murphy KT, Mahajan S, Mahajan Y, Levin C, Blum K. Theorizing that Psychedelic Assisted Therapy May Play a Role in the Treatment of Trauma-Induced Personality Disorders. JOURNAL OF ADDICTION PSYCHIATRY 2024; 8:161-165. [PMID: 39634920 PMCID: PMC11616086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Borderline personality disorder (BPD) and post-traumatic stress disorder (PTSD) share overlapping neurobiological mechanisms particularly reward deficiency and stress-like anti-reward processes. And so, BPD may be reclassified as a "traumatic personality stress disorder" (TPSD) with ensuing common therapeutic strategies that may stabilize dopaminergic reward function such as psychedelic-assisted therapy. Integrated therapeutic strategies may be further supported by genetic studies aimed at assessing similarities between the two therapeutic entities. In this perspective we theorize that psychedelic assisted therapy (PAT) may play a role in the treatment of trauma induced personality disorders. This study identifies PAT as a pathway for treating both BPD and PTSD, proposing that reframing BPD as TPSD could lead to more effective, personalized interventions, ultimately improving the quality of life for those affected by trauma. Such a reclassification might also mitigate stigma, enhance our understanding of the underlying mechanisms, and optimize therapeutic interventions for a broader range of diagnostic categories characterized by anhedonia, negative affective states, hypervigilance, and dissociation.
Collapse
Affiliation(s)
- Gianni Martire
- Biophysics Research in Advanced Interdisciplinary Neuroscience at Applied Physics, New York, USA
| | | | - David Baron
- Division of Addiction Research, Therapy and Education, Center for Sports, Exercise and Mental Health, Western Health Sciences, Pomona, USA
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, USA
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, USA
| | - Kai-Uwe Lewandowski
- Division of Personalized Pain Therapy Research & Education, Center for Advanced Spine Care of Southern Arizona, Tucson, USA
- Department of Orthopaedics, Fundación Universitaria Sanitas Bogotá D.C. Colombia
- Department of Orthopaedics, Universidade Federal do Estado do Rio de Janeiro, Brazil
| | - Catherine A. Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, USA
| | - Alireza Sharafshah
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Igor Elman
- Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Panayotis K. Thanos
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, USA
| | - Edward J. Modestino
- Brain & Behavior Laboratory, Department of Psychology, Curry College , Milton, MA., USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, Texas Tech University Health Sciences, School of Medicine, Lubbock, USA
| | - Albert Pinhasov
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Milan Makale
- Department of Radiation Oncology, University of California San Diego, La Jolla, CA, USA
| | - A. Kenison Roy
- Department of Psychiatry, Tulane University School of Medicine, New Orleans, LA., USA
| | - Keerthy Sunder
- Karma Doctors & Karma TMS, and Suder Foundation, Palm Springs, CA, USA
- Department of Medicine, University of California, Riverside School of Medicine, Riverside, CA, USA
| | - Kevin T. Murphy
- Division of personalized neuromodulation, Peak Logic, LLC., Del Mar, CA., USA
| | - Shaurya Mahajan
- Karma Doctors & Karma TMS, and Suder Foundation, Palm Springs, CA, USA
| | - Yatharth Mahajan
- Karma Doctors & Karma TMS, and Suder Foundation, Palm Springs, CA, USA
| | - Chynna Levin
- The Blum Neurogenetic & Behavioral Institute, LLC., Austin, USA
| | - Kenenth Blum
- Division of Addiction Research, Therapy and Education, Center for Sports, Exercise and Mental Health, Western Health Sciences, Pomona, USA
- Division of Personalized Pain Therapy Research & Education, Center for Advanced Spine Care of Southern Arizona, Tucson, USA
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, Israel
- Karma Doctors & Karma TMS, and Suder Foundation, Palm Springs, CA, USA
- The Blum Neurogenetic & Behavioral Institute, LLC., Austin, USA
| |
Collapse
|
5
|
Barrios KP, Connolly DJ, Ferris JA, Maier LJ, Barratt MJ, Winstock AR, Puljević C, Gilchrist G. Ketamine use in a large global sample: Characteristics, patterns of use and emergency medical treatment. J Psychopharmacol 2024:2698811241273850. [PMID: 39420535 DOI: 10.1177/02698811241273850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
BACKGROUND Ketamine's popularity has surged globally in the past decade, especially among young men. Emergency department visits due to its toxicity remain relatively rare, often linked to co-occurring use of other substances. AIMS Using data from the Global Drug Survey (GDS) 2018, this study explored the correlates associated with lifetime and past-year ketamine use, and estimated the socio-demographic characteristics, usage patterns and experiences of respondents seeking emergency medical treatment (EMT) after ketamine use. METHODS Secondary analysis of GDS 2018, an online cross-sectional survey on drug use patterns conducted between November 2017 and January 2018. RESULTS The survey received 130,761 valid responses, with 5.93% reporting lifetime ketamine use, of which 57.70% used ketamine within the past year. Predominantly, respondents were from Germany, England and Denmark. Within the past year, 8.55% met the criteria for ketamine dependence. Respondents who used ketamine in their lifetime tended to be young (mean (x̄) = 27.37 years), men, heterosexual and of white ethnicity. Younger age (x̄ = 24.84 years), gay sexual orientation, student status, past-year use of other drugs and no lifetime mental health diagnosis were associated with past-year ketamine use. Among 4477 respondents reporting past-year ketamine use, 120 adverse events were reported, with less than 0.10% prompting EMT seeking. CONCLUSION The study reveals frequent ketamine use but low harm occurrence, underscoring the complex interplay between ketamine use, substance use and dependence, and related factors. This underscores the need to reassess EMT priorities, implement tailored harm reduction strategies and incorporate comprehensive screening for addressing ketamine and substance dependence challenges.
Collapse
Affiliation(s)
- Karen P Barrios
- National Addiction Center, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Dean J Connolly
- National Addiction Center, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Jason A Ferris
- Faculty of Medicine, Center for Health Services Research, The University of Queensland, Brisbane, QLD, Australia
| | - Larissa J Maier
- Department of Clinical Pharmacy, University of California, San Francisco, CA, USA
| | - Monica J Barratt
- Social Equity Research Center and Digital Ethnography Research Center, RMIT University, Melbourne, VIC, Australia
- National Drug and Alcohol Research Center, UNSW Sydney, Sydney, NSW, Australia
| | - Adam R Winstock
- Global Drug Survey, London, UK
- Institute of Epidemiology and Health Care, University College London, London, UK
| | - Cheneal Puljević
- Faculty of Medicine, School of Public Health, The University of Queensland, Brisbane, QLD, Australia
| | - Gail Gilchrist
- National Addiction Center, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
6
|
Kajumba MM, Kakooza-Mwesige A, Nakasujja N, Koltai D, Canli T. Treatment-resistant depression: molecular mechanisms and management. MOLECULAR BIOMEDICINE 2024; 5:43. [PMID: 39414710 PMCID: PMC11485009 DOI: 10.1186/s43556-024-00205-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/03/2024] [Indexed: 10/18/2024] Open
Abstract
Due to the heterogeneous nature of depression, the underlying etiological mechanisms greatly differ among individuals, and there are no known subtype-specific biomarkers to serve as precise targets for therapeutic efficacy. The extensive research efforts over the past decades have not yielded much success, and the currently used first-line conventional antidepressants are still ineffective for close to 66% of patients. Most clinicians use trial-and-error treatment approaches, which seem beneficial to only a fraction of patients, with some eventually developing treatment resistance. Here, we review evidence from both preclinical and clinical studies on the pathogenesis of depression and antidepressant treatment response. We also discuss the efficacy of the currently used pharmacological and non-pharmacological approaches, as well as the novel emerging therapies. The review reveals that the underlying mechanisms in the pathogenesis of depression and antidepressant response, are not specific, but rather involve an interplay between various neurotransmitter systems, inflammatory mediators, stress, HPA axis dysregulation, genetics, and other psycho-neurophysiological factors. None of the current depression hypotheses sufficiently accounts for the interactional mechanisms involved in both its etiology and treatment response, which could partly explain the limited success in discovering efficacious antidepressant treatment. Effective management of treatment-resistant depression (TRD) requires targeting several interactional mechanisms, using subtype-specific and/or personalized therapeutic modalities, which could, for example, include multi-target pharmacotherapies in augmentation with psychotherapy and/or other non-pharmacological approaches. Future research guided by interaction mechanisms hypotheses could provide more insights into potential etiologies of TRD, precision biomarker targets, and efficacious therapeutic modalities.
Collapse
Affiliation(s)
- Mayanja M Kajumba
- Department of Mental Health and Community Psychology, Makerere University, P. O. Box 7062, Kampala, Uganda.
| | - Angelina Kakooza-Mwesige
- Department of Pediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
- Department of Pediatrics and Child Health, Mulago National Referral Hospital, Kampala, Uganda
| | - Noeline Nakasujja
- Department of Psychiatry, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Deborah Koltai
- Duke Division of Global Neurosurgery and Neurology, Department of Neurosurgery, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, USA
| | - Turhan Canli
- Department of Psychology, Stony Brook University, New York, USA
- Department of Psychiatry, Stony Brook University, New York, USA
| |
Collapse
|
7
|
Yip E, Fleck D. The Use of Prophylactic Ketamine to Mitigate Postoperative Depressive Symptoms: A Systematic Review. J Perianesth Nurs 2024:S1089-9472(24)00210-7. [PMID: 39365203 DOI: 10.1016/j.jopan.2024.05.026] [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: 09/06/2023] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE Depression is predicted to be the leading burden of disease worldwide by 2030 with a prevalence of 10% to 60% in the surgical population. Depressive symptoms in the perioperative population are associated with a myriad of grave complications, including higher morbidity and mortality. This systematic review aims to determine whether a single dose of intravenous ketamine can be used prophylactically as a routine resilience-enhancing agent in all high-risk adult patients undergoing surgery to mitigate depressive symptoms postoperatively by appraising evidence of existing literature. DESIGN An evidence-based systematic review. METHODS The databases PubMed, CINAHL, and Scopus were searched, adhering to the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Search and Medical Subject Headings (MeSH) terms used: "ketamine," "surgical procedures," "surgical," "surgery," "depression," and "depressive disorder." FINDINGS In 9 randomized controlled trials (RCTs), bolus ketamine doses of 0.1 mg/kg to 1 mg/kg were administered intraoperatively or in the postanesthesia care unit. The primary outcome was the subjective feelings of depression as evaluated by different validated depression screening tools and seven RCTs observed statistically significant findings between the control and intervention (ketamine) groups with postoperative depression scores. Two RCTs did not find a significant change in depression scores. CONCLUSIONS Research on the use of prophylactic ketamine as an antidepressant in the perioperative population offers insight into changing routine practices and can shape how mental health is viewed in hospitals. Further research is warranted on the safety profile of ketamine, and risk stratification with careful consideration of baseline depressive symptoms, timing, and its use across a wider variety of surgical subspecialties.
Collapse
Affiliation(s)
- Eleanor Yip
- University of Pennsylvania School of Nursing, Philadelphia, PA.
| | - Desiree Fleck
- University of Pennsylvania School of Nursing, Philadelphia, PA
| |
Collapse
|
8
|
Mandal G, Kirkpatrick M, Alboni S, Mariani N, Pariante CM, Borsini A. Ketamine Prevents Inflammation-Induced Reduction of Human Hippocampal Neurogenesis via Inhibiting the Production of Neurotoxic Metabolites of the Kynurenine Pathway. Int J Neuropsychopharmacol 2024; 27:pyae041. [PMID: 39297528 PMCID: PMC11450635 DOI: 10.1093/ijnp/pyae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Understanding the precise mechanisms of ketamine is crucial for replicating its rapid antidepressant effects without inducing psychomimetic changes. Here, we explore whether the antidepressant-like effects of ketamine enantiomers are underscored by protection against cytokine-induced reductions in hippocampal neurogenesis and activation of the neurotoxic kynurenine pathway in our well-established in vitro model of depression in a dish. METHODS We used the fetal hippocampal progenitor cell line (HPC0A07/03C) to investigate ketamine's impact on cytokine-induced reductions in neurogenesis in vitro. Cells were treated with interleukin- 1beta (IL-1b) (10 ng/mL) or IL-6 (50 pg/mL), alone or in combination with ketamine enantiomers arketamine (R-ketamine, 400 nM) or esketamine (S-ketamine, 400 nM) or antidepressants sertraline (1 mM) or venlafaxine (1 mM). RESULTS Resembling the effect of antidepressants, both ketamine enantiomers prevented IL-1b- and IL-6-induced reduction in neurogenesis and increase in apoptosis. This was mediated by inhibition of IL-1b-induced production of IL-2 and IL-13 by R-ketamine and of IL-1b-induced tumor necrosis factor-alpha by S-ketamine. Likewise, R-ketamine inhibited IL-6-induced production of IL-13, whereas S-ketamine inhibited IL-6-induced IL-1b and IL-8. Moreover, both R- and S-ketamine prevented IL-1b-induced increases in indoleamine 2,3-dioxygenase expression as well as kynurenine production, which in turn was shown to mediate the detrimental effects of IL-1b on neurogenesis and apoptosis. In contrast, neither R- nor S-ketamine prevented IL-6-induced kynurenine pathway activation. CONCLUSIONS Results suggest that R- and S-ketamine have pro-neurogenic and anti-inflammatory properties; however, this is mediated by inhibition of the kynurenine pathway only in the context of IL-1b. Overall, this study enhances our understanding of the mechanisms underlying ketamine's antidepressant effects in the context of different inflammatory phenotypes, ultimately leading to the development of more effective, personalized therapeutic approaches for patients suffering from depression.
Collapse
Affiliation(s)
- Gargi Mandal
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King’s College London, UK
| | - Madeline Kirkpatrick
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King’s College London, UK
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicole Mariani
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King’s College London, UK
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King’s College London, UK
| | - Alessandra Borsini
- Stress, Psychiatry and Immunology Laboratory, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, King’s College London, UK
| |
Collapse
|
9
|
Wen W, Wenjing Z, Xia X, Duan X, Zhang L, Duomao L, Zeyou Q, Wang S, Gao M, Liu C, Li H, Ma J. Efficacy of ketamine versus esketamine in the treatment of perioperative depression: A review. Pharmacol Biochem Behav 2024; 242:173773. [PMID: 38806116 DOI: 10.1016/j.pbb.2024.173773] [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: 10/31/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Depression is a significant factor contributing to postoperative occurrences, and patients diagnosed with depression have a higher risk for postoperative complications. Studies on cardiovascular surgery extensively addresses this concern. Several studies report that people who undergo coronary artery bypass graft surgery have a 20% chance of developing postoperative depression. A retrospective analysis of medical records spanning 21 years, involving 817 patients, revealed that approximately 40% of individuals undergoing coronary artery bypass grafting (CABG) were at risk of perioperative depression. Patients endure prolonged suffering from illness because each attempt with standard antidepressants requires several weeks to be effective. In addition, multi-drug combination adjuvants or combination medication therapy may alleviate symptoms for some individuals, but they also increase the risk of side effects. Conventional antidepressants primarily modulate the monoamine system, whereas different therapies target the serotonin, norepinephrine, and dopamine systems. Esketamine is a fast-acting antidepressant with high efficacy. Esketamine is the S-enantiomer of ketamine, a derivative of phencyclidine developed in 1956. Esketamine exerts its effect by targeting the glutaminergic system the glutaminergic system. In this paper, we discuss the current depression treatment strategies with a focus on the pharmacology and mechanism of action of esketamine. In addition, studies reporting use of esketamine to treat perioperative depressive symptoms are reviwed, and the potential future applications of the drug are presented.
Collapse
Affiliation(s)
- Wen Wen
- Beijing Anzhen Hospital, Capital Medical University
| | - Zhao Wenjing
- Beijing Anzhen Hospital, Capital Medical University
| | - Xing Xia
- Beijing Anzhen Hospital, Capital Medical University
| | | | - Liang Zhang
- Beijing Anzhen Hospital, Capital Medical University
| | - Lin Duomao
- Beijing Anzhen Hospital, Capital Medical University
| | - Qi Zeyou
- Beijing Anzhen Hospital, Capital Medical University
| | - Sheng Wang
- Beijing Anzhen Hospital, Capital Medical University
| | - Mingxin Gao
- Beijing Anzhen Hospital, Capital Medical University
| | | | - Haiyang Li
- Beijing Anzhen Hospital, Capital Medical University.
| | - Jun Ma
- Beijing Anzhen Hospital, Capital Medical University.
| |
Collapse
|
10
|
Ahmadi J, Mansoori A, Mosavat SH, Bazrafshan A. Comparison of ketamine with buprenorphine as adjunctive therapy in the treatment of comorbid major depressive disorder and opium use disorders: A randomized controlled trial. Int J Psychiatry Med 2024; 59:521-535. [PMID: 38140979 DOI: 10.1177/00912174231225087] [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] [Indexed: 12/24/2023]
Abstract
BACKGROUND Comorbid major depressive disorder (MDD) and opium use disorder (OUD) are known to increase the risk of suicide. The purpose of this study was to compare the efficacy and safety of adjunctive therapy with either ketamine or buprenorphine in patients with comorbid MDD and OUD. METHODS This was a randomized double-blind controlled trial in adults admitted to a hospital in Iran. Sixty-six participants were enrolled and received either ketamine or buprenorphine, along with current antidepressant therapy. The primary outcome was change in depressive symptoms assessed using the Beck Depression Inventory (BDI) after 2 hours, 24 hours, and 7 days following initiation of treatment. Secondary outcomes included changes in suicidal ideation, evaluated by the Beck Scale for Suicidal Ideation (BSSI). RESULTS Both groups experienced a significant decrease in the severity of depression following the interventions (p < .05). However, there was no significant difference in the between-group comparison (p > .05). Both groups also exhibited a significant reduction in suicidal ideation compared to before the study, with the decrease in severity being over 85% in both groups (p < .05). CONCLUSION Both ketamine and buprenorphine appear to be equally effective in reducing symptoms of depression and suicidal ideation among individuals with MDD and OUD.
Collapse
Affiliation(s)
- Jamshid Ahmadi
- Substance Abuse Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- IMCES (Institute for Multicultural Counseling and Educational Services), Los Angeles, CA, USA
| | - Arash Mansoori
- Research Center for Psychiatry and Behavioral Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Hamdollah Mosavat
- Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Bazrafshan
- Substance Abuse Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Yates JR. Pharmacological Treatments for Methamphetamine Use Disorder: Current Status and Future Targets. Subst Abuse Rehabil 2024; 15:125-161. [PMID: 39228432 PMCID: PMC11370775 DOI: 10.2147/sar.s431273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
The illicit use of the psychostimulant methamphetamine (METH) is a major concern, with overdose deaths increasing substantially since the mid-2010s. One challenge to treating METH use disorder (MUD), as with other psychostimulant use disorders, is that there are no available pharmacotherapies that can reduce cravings and help individuals achieve abstinence. The purpose of the current review is to discuss the molecular targets that have been tested in assays measuring the physiological, the cognitive, and the reinforcing effects of METH in both animals and humans. Several drugs show promise as potential pharmacotherapies for MUD when tested in animals, but fail to produce long-term changes in METH use in dependent individuals (eg, modafinil, antipsychotic medications, baclofen). However, these drugs, plus medications like atomoxetine and varenicline, may be better served as treatments to ameliorate the psychotomimetic effects of METH or to reverse METH-induced cognitive deficits. Preclinical studies show that vesicular monoamine transporter 2 inhibitors, metabotropic glutamate receptor ligands, and trace amine-associated receptor agonists are efficacious in attenuating the reinforcing effects of METH; however, clinical studies are needed to determine if these drugs effectively treat MUD. In addition to screening these compounds in individuals with MUD, potential future directions include increased emphasis on sex differences in preclinical studies and utilization of pharmacogenetic approaches to determine if genetic variances are predictive of treatment outcomes. These future directions can help lead to better interventions for treating MUD.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA
| |
Collapse
|
12
|
Park I, Choi M, Kim J, Jang S, Kim D, Kim J, Choe Y, Geum D, Yu SW, Choi JW, Moon C, Choe HK, Son GH, Kim K. Role of the circadian nuclear receptor REV-ERBα in dorsal raphe serotonin synthesis in mood regulation. Commun Biol 2024; 7:998. [PMID: 39147805 PMCID: PMC11327353 DOI: 10.1038/s42003-024-06647-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 07/29/2024] [Indexed: 08/17/2024] Open
Abstract
Affective disorders are frequently associated with disrupted circadian rhythms. The existence of rhythmic secretion of central serotonin (5-hydroxytryptamine, 5-HT) pattern has been reported; however, the functional mechanism underlying the circadian control of 5-HTergic mood regulation remains largely unknown. Here, we investigate the role of the circadian nuclear receptor REV-ERBα in regulating tryptophan hydroxylase 2 (Tph2), the rate-limiting enzyme of 5-HT synthesis. We demonstrate that the REV-ERBα expressed in dorsal raphe (DR) 5-HTergic neurons functionally competes with PET-1-a nuclear activator crucial for 5-HTergic neuron development. In mice, genetic ablation of DR 5-HTergic REV-ERBα increases Tph2 expression, leading to elevated DR 5-HT levels and reduced depression-like behaviors at dusk. Further, pharmacological manipulation of the mice DR REV-ERBα activity increases DR 5-HT levels and affects despair-related behaviors. Our findings provide valuable insights into the molecular and cellular link between the circadian rhythm and the mood-controlling DR 5-HTergic systems.
Collapse
Affiliation(s)
- Inah Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Mijung Choi
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Jeongah Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Sangwon Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Doyeon Kim
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Jihoon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Dongho Geum
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Seong-Woon Yu
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Ji-Woong Choi
- Department of Electrical Engineering and Computer Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Cheil Moon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Han Kyoung Choe
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Gi Hoon Son
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Legal Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Kyungjin Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| |
Collapse
|
13
|
Guldager MB, Chaves Filho AM, Biojone C, Joca S. Therapeutic potential of cannabidiol in depression. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 177:251-293. [PMID: 39029987 DOI: 10.1016/bs.irn.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Major depressive disorder (MDD) is a widespread and debilitating condition affecting a significant portion of the global population. Traditional treatment for MDD has primarily involved drugs that increase brain monoamines by inhibiting their uptake or metabolism, which is the basis for the monoaminergic hypothesis of depression. However, these treatments are only partially effective, with many patients experiencing delayed responses, residual symptoms, or complete non-response, rendering the current view of the hypothesis as reductionist. Cannabidiol (CBD) has shown promising results in preclinical models and human studies. Its mechanism is not well-understood, but may involve monoamine and endocannabinoid signaling, control of neuroinflammation and enhanced neuroplasticity. This chapter will explore CBD's effects in preclinical and clinical studies, its molecular mechanisms, and its potential as a treatment for MDD.
Collapse
Affiliation(s)
- Matti Bock Guldager
- Department of Biomedicine, Health Faculty, Aarhus University, Aarhus, Denmark; Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Health Faculty, Aarhus University, Aarhus, Denmark
| | | | - Caroline Biojone
- Department of Biomedicine, Health Faculty, Aarhus University, Aarhus, Denmark; Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Health Faculty, Aarhus University, Aarhus, Denmark
| | - Sâmia Joca
- Department of Biomedicine, Health Faculty, Aarhus University, Aarhus, Denmark; Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Health Faculty, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
14
|
Gent EM, Bryan JW, Cleary MA, Clarke TI, Holmwood HD, Nassereddine RO, Salway C, Depla S, Statton S, Krecké J, Morgan CJA. Esketamine combined with a mindfulness-based intervention for individuals with alcohol problems. J Psychopharmacol 2024; 38:541-550. [PMID: 38863284 PMCID: PMC11179319 DOI: 10.1177/02698811241254834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
BACKGROUND Alcohol use disorder (AUD) is a major public health issue, posing harmful consequences for individuals and society. Recent advances in addiction research have highlighted the therapeutic potential of ketamine-assisted therapy for AUD. However, the exact mechanisms underlying its effectiveness remain unknown. AIMS This double-blind, pilot study aimed to investigate esketamine combined with mindfulness-based intervention (MBI) to examine whether esketamine enhances engagement in MBI for individuals with alcohol misuse problems and whether enhanced engagement has any impact on alcohol-related outcomes. METHODS In all, 28 individuals with alcohol problems were randomly assigned to receive sublingual esketamine hydrochloride (AWKN002: 115.1 mg) or vitamin C (placebo) in an oral thin film and took part in 2 weeks of daily MBI. Participants were assessed on various self-report measures, including mindfulness, engagement in MBI (physical and psychological), alcohol cravings and consumption. RESULTS Esketamine enhanced psychological engagement with a daily MBI, compared to placebo, and led to transient decreases in alcohol cravings. Esketamine also resulted in significantly greater mystical experiences and dissociative states compared to placebo. CONCLUSIONS The findings suggest that esketamine may improve treatment outcomes when combined with mindfulness-based therapies through its ability to increase engagement with meditative practice.
Collapse
Affiliation(s)
- Emily M Gent
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Joshua W Bryan
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Maisy A Cleary
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Tegan I Clarke
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Harry D Holmwood
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Rania O Nassereddine
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Chris Salway
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Simon Depla
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Sarah Statton
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Joy Krecké
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| | - Celia JA Morgan
- Psychopharmacology and Addiction Research Centre, Haighton Research Centre, University of Exeter St Luke’s Campus, Exeter, UK
| |
Collapse
|
15
|
Tizabi Y, Getachew B, Hauser SR, Tsytsarev V, Manhães AC, da Silva VDA. Role of Glial Cells in Neuronal Function, Mood Disorders, and Drug Addiction. Brain Sci 2024; 14:558. [PMID: 38928557 PMCID: PMC11201416 DOI: 10.3390/brainsci14060558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Mood disorders and substance use disorder (SUD) are of immense medical and social concern. Although significant progress on neuronal involvement in mood and reward circuitries has been achieved, it is only relatively recently that the role of glia in these disorders has attracted attention. Detailed understanding of the glial functions in these devastating diseases could offer novel interventions. Here, following a brief review of circuitries involved in mood regulation and reward perception, the specific contributions of neurotrophic factors, neuroinflammation, and gut microbiota to these diseases are highlighted. In this context, the role of specific glial cells (e.g., microglia, astroglia, oligodendrocytes, and synantocytes) on phenotypic manifestation of mood disorders or SUD are emphasized. In addition, use of this knowledge in the potential development of novel therapeutics is touched upon.
Collapse
Affiliation(s)
- Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA;
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA;
| | - Sheketha R. Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Vassiliy Tsytsarev
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Alex C. Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, IBRAG, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-170, RJ, Brazil
| | - Victor Diogenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-100, BA, Brazil;
| |
Collapse
|
16
|
Kovoor JG, Santhosh S, Stretton B, Tan S, Gouldooz H, Moorthy S, Pietris J, Hannemann C, Yu LK, Johnson R, Reddi BA, Gupta AK, Wagner M, Page GJ, Kovoor P, Bastiampillai T, Maddocks I, Perry SW, Wong ML, Licinio J, Bacchi S. Near-death experiences after cardiac arrest: a scoping review. DISCOVER MENTAL HEALTH 2024; 4:19. [PMID: 38806961 PMCID: PMC11133272 DOI: 10.1007/s44192-024-00072-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 05/24/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND This scoping review aimed to characterise near-death experiences in the setting of cardiac arrest, a phenomenon that is poorly understood and may have clinical consequences. METHOD PubMed/MEDLINE was searched to 23 July 2023 for prospective studies describing near-death experiences in cardiac arrest. PRISMA-ScR guidelines were adhered to. Qualitative and quantitative data were synthesised. Meta-analysis was precluded due to data heterogeneity. RESULTS 60 records were identified, of which 11 studies involving interviews were included from various countries. Sample size ranged from 28-344, and proportion of female patients (when reported) was 0-50%, with mean age (when reported) ranging 54-64 years. Comorbidities and reasons for cardiac arrest were heterogeneously reported. Incidence of near-death experiences in the included studies varied from 6.3% to 39.3%; with variation between in-hospital (6.3-39.3%) versus out-of-hospital (18.9-21.2%) cardiac arrest. Individual variables regarding patient characteristics demonstrated statistically significant association with propensity for near-death experiences. Reported content of near-death experiences tended to reflect the language of the questionnaires used, rather than the true language used by individual study participants. Three studies conducted follow-up, and all suggested a positive life attitude change, however one found significantly higher 30-day all-cause mortality in patients with near-death experiences versus those without, in non-controlled analysis. CONCLUSIONS From prospective studies that have investigated the phenomenon, near-death experiences may occur in as frequent as over one-third of patients with cardiac arrest. Lasting effects may follow these events, however these could also be confounded by clinical characteristics.
Collapse
Affiliation(s)
- Joshua G Kovoor
- University of Adelaide, Adelaide, South Australia, Australia
- Ballarat Base Hospital, Ballarat, VIC, Australia
- Royal Adelaide Hospital, Adelaide, South Australia, Australia
- Health and Information, Adelaide, South Australia, Australia
- Heart of the Nation, Sydney, NSW, Australia
| | | | - Brandon Stretton
- University of Adelaide, Adelaide, South Australia, Australia
- Royal Adelaide Hospital, Adelaide, South Australia, Australia
- Health and Information, Adelaide, South Australia, Australia
| | - Sheryn Tan
- University of Adelaide, Adelaide, South Australia, Australia
- Health and Information, Adelaide, South Australia, Australia
| | - Hasti Gouldooz
- Gold Coast University Hospital, Gold Coast, QLD, Australia
| | | | | | - Christopher Hannemann
- Ballarat Base Hospital, Ballarat, VIC, Australia
- Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Long Kiu Yu
- University of Adelaide, Adelaide, South Australia, Australia
| | - Rhys Johnson
- University of Adelaide, Adelaide, South Australia, Australia
| | - Benjamin A Reddi
- University of Adelaide, Adelaide, South Australia, Australia
- Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Aashray K Gupta
- University of Adelaide, Adelaide, South Australia, Australia
- Health and Information, Adelaide, South Australia, Australia
- Gold Coast University Hospital, Gold Coast, QLD, Australia
| | - Morganne Wagner
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | | | - Pramesh Kovoor
- Heart of the Nation, Sydney, NSW, Australia
- Westmead Hospital, Sydney, NSW, Australia
- University of Sydney, Sydney, NSW, Australia
| | - Tarun Bastiampillai
- Flinders University, Adelaide, South Australia, Australia
- Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Ian Maddocks
- Australasian Chapter of Palliative Medicine, Royal Australasian College of Physicians, Adelaide, South Australia, Australia
| | - Seth W Perry
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ma-Li Wong
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Julio Licinio
- State University of New York Upstate Medical University, Syracuse, NY, USA
- Flinders University, Adelaide, South Australia, Australia
| | - Stephen Bacchi
- Health and Information, Adelaide, South Australia, Australia.
- Flinders University, Adelaide, South Australia, Australia.
- Lyell McEwin Hospital, Adelaide, South Australia, Australia.
| |
Collapse
|
17
|
Glavonic E, Dragic M, Mitic M, Aleksic M, Lukic I, Ivkovic S, Adzic M. Ketamine's Amelioration of Fear Extinction in Adolescent Male Mice Is Associated with the Activation of the Hippocampal Akt-mTOR-GluA1 Pathway. Pharmaceuticals (Basel) 2024; 17:669. [PMID: 38931336 PMCID: PMC11206546 DOI: 10.3390/ph17060669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/29/2024] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Fear-related disorders, including post-traumatic stress disorder (PTSD), and anxiety disorders are pervasive psychiatric conditions marked by persistent fear, stemming from its dysregulated acquisition and extinction. The primary treatment for these disorders, exposure therapy (ET), relies heavily on fear extinction (FE) principles. Adolescence, a vulnerable period for developing psychiatric disorders, is characterized by neurobiological changes in the fear circuitry, leading to impaired FE and increased susceptibility to relapse following ET. Ketamine, known for relieving anxiety and reducing PTSD symptoms, influences fear-related learning processes and synaptic plasticity across the fear circuitry. Our study aimed to investigate the effects of ketamine (10 mg/kg) on FE in adolescent male C57 BL/6 mice at the behavioral and molecular levels. We analyzed the protein and gene expression of synaptic plasticity markers in the hippocampus (HPC) and prefrontal cortex (PFC) and sought to identify neural correlates associated with ketamine's effects on adolescent extinction learning. Ketamine ameliorated FE in the adolescent males, likely affecting the consolidation and/or recall of extinction memory. Ketamine also increased the Akt and mTOR activity and the GluA1 and GluN2A levels in the HPC and upregulated BDNF exon IV mRNA expression in the HPC and PFC of the fear-extinguished mice. Furthermore, ketamine increased the c-Fos expression in specific brain regions, including the ventral HPC (vHPC) and the left infralimbic ventromedial PFC (IL vmPFC). Providing a comprehensive exploration of ketamine's mechanisms in adolescent FE, our study suggests that ketamine's effects on FE in adolescent males are associated with the activation of hippocampal Akt-mTOR-GluA1 signaling, with the vHPC and the left IL vmPFC as the proposed neural correlates.
Collapse
Affiliation(s)
- Emilija Glavonic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Milorad Dragic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11158 Belgrade, Serbia
| | - Milos Mitic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Minja Aleksic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Iva Lukic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11351 Belgrade, Serbia; (E.G.); (M.D.); (M.M.); (M.A.); (I.L.); (S.I.)
| |
Collapse
|
18
|
Zhou L, Duan J. The role of NMDARs in the anesthetic and antidepressant effects of ketamine. CNS Neurosci Ther 2024; 30:e14464. [PMID: 37680076 PMCID: PMC11017467 DOI: 10.1111/cns.14464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND As a phencyclidine (PCP) analog, ketamine can generate rapid-onset and substantial anesthetic effects. Contrary to traditional anesthetics, ketamine is a dissociative anesthetic and can induce loss of consciousness in patients. Recently, the subanaesthetic dose of ketamine was found to produce rapid-onset and lasting antidepressant effects. AIM However, how different concentrations of ketamine can induce diverse actions remains unclear. Furthermore, the molecular mechanisms underlying the NMDAR-mediated anesthetic and antidepressant effects of ketamine are not fully understood. METHOD In this review, we have introduced ketamine and its metabolism, summarized recent advances in the molecular mechanisms underlying NMDAR inhibition in the anesthetic and antidepressant effects of ketamine, explored the possible functions of NMDAR subunits in the effects of ketamine, and discussed the future directions of ketamine-based anesthetic and antidepressant drugs. RESULT Both the anesthetic and antidepressant effects of ketamine were thought to be mediated by N-methyl-D-aspartate receptor (NMDAR) inhibition. CONCLUSION The roles of NMDARs have been extensively studied in the anaesthetic effects of ketamine. However, the roles of NMDARs in antidepressant effects of ketamine are complicated and controversial.
Collapse
Affiliation(s)
- Liang Zhou
- Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Jingjing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of MedicineSunYat‐sen UniversityGuangzhouChina
| |
Collapse
|
19
|
Sajid S, Galfalvy HC, Keilp JG, Burke AK, Mann JJ, Grunebaum MF. Acute Dissociation and Ketamine's Antidepressant and Anti-Suicidal Ideation Effects in a Midazolam-Controlled Trial. Int J Neuropsychopharmacol 2024; 27:pyae017. [PMID: 38573154 PMCID: PMC11053360 DOI: 10.1093/ijnp/pyae017] [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: 01/02/2024] [Accepted: 04/03/2024] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVE We sought to explore relationships of acute dissociative effects of intravenous ketamine with change in depression and suicidal ideation and with plasma metabolite levels in a randomized, midazolam-controlled trial. METHODS Data from a completed trial in suicidal, depressed participants (n = 40) randomly assigned to ketamine was used to examine relationships between ketamine treatment-emergent dissociative and psychotomimetic symptoms with pre/post-infusion changes in suicidal ideation and depression severity. Nonparametric correlational statistics were used. These methods were also used to explore associations between dissociative or psychotomimetic symptoms and blood levels of ketamine and metabolites in a subset of participants (n = 28) who provided blood samples immediately post-infusion. RESULTS Neither acute dissociative nor psychotomimetic effects of ketamine were associated with changes in suicidal ideation or depressive symptoms from pre- to post-infusion. Norketamine had a trend-level, moderate inverse correlation with dissociative symptoms on Day 1 post-injection (P = .064; P =.013 removing 1 outlier). Dehydronorketamine correlated with Clinician-Administered Dissociative States Scale scores at 40 minutes (P = .034), 230 minutes (P = .014), and Day 1 (P = .012). CONCLUSION We did not find evidence that ketamine's acute, transient dissociative, or psychotomimetic effects are associated with its antidepressant or anti-suicidal ideation actions. The correlation of higher plasma norketamine with lower dissociative symptoms on Day 1 post-treatment suggests dissociation may be more an effect of the parent drug.
Collapse
Affiliation(s)
- Sumra Sajid
- New York State Psychiatric Institute, New York, New York, USA
| | - Hanga C Galfalvy
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, New York, USA
| | - John G Keilp
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, New York, USA
| | - Ainsley K Burke
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, New York, USA
| | - J John Mann
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, New York, USA
| | - Michael F Grunebaum
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, New York, USA
| |
Collapse
|
20
|
Yan R, Marshall T, Khullar A, Nagle T, Knowles J, Malkin M, Chubbs B, Swainson J. Patient-reported outcomes on sleep quality and circadian rhythm during treatment with intravenous ketamine for treatment-resistant depression. Ther Adv Psychopharmacol 2024; 14:20451253241231264. [PMID: 38440104 PMCID: PMC10910882 DOI: 10.1177/20451253241231264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/18/2024] [Indexed: 03/06/2024] Open
Abstract
Background Intravenous (IV) ketamine is a rapid acting antidepressant used primarily for treatment-resistant depression (TRD). It has been suggested that IV ketamine's rapid antidepressant effects may be partially mediated via improved sleep and changes to the circadian rhythm. Objectives This study explores IV ketamine's association with changes in patient-reported sleep quality and circadian rhythm in an adult population with TRD. Methods Adult patients (18-64 years) with TRD scheduled for IV ketamine treatment were recruited to complete patient rated outcomes measures on sleep quality using the Pittsburgh Sleep Quality Index (PSQI) and circadian rhythm using the Morningness-Eveningness Questionnaire (MEQ). Over a 4-week course of eight ketamine infusions, reports were obtained at baseline (T0), prior to second treatment (T1), prior to fifth treatment (T2), and 1 week after eighth treatment (T3). Results Forty participants with TRD (mean age = 42.8, 45% male) were enrolled. Twenty-nine (72.5%) had complete follow-up data. Paired t tests revealed statistically significant improvements at the end of treatment in sleep quality (PSQI) (p = 0.003) and depressive symptoms (Clinically Useful Depression Outcome Scale-Depression, p < 0.001) while circadian rhythm (MEQ) shifted earlier (p = 0.007). The PSQI subscale components of sleep duration (p = 0.008) and daytime dysfunction (p = 0.001) also improved. In an exploratory post hoc analysis, ketamine's impact on sleep quality was more prominent in patients with mixed features, while its chronobiotic effect was prominent in those without mixed features. Conclusion IV ketamine may improve sleep quality and advance circadian rhythm in individuals with TRD. Effects may differ in individuals with mixed features of depression as compared to those without. Since this was a small uncontrolled study, future research is warranted.
Collapse
Affiliation(s)
- Raymond Yan
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Atul Khullar
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Travis Nagle
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Jake Knowles
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Mai Malkin
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Brittany Chubbs
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada
| | - Jennifer Swainson
- Cabrini Center, 3rd Floor, 16811-88 Ave NW, Edmonton, AB, Canada T5R 5YR
- Department of Psychiatry, University of Alberta, Canada
- Misericordia Community Hospital, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Famuła A, Radoszewski J, Czerwiec T, Sobiś J, Więckiewicz G. Ketamine in Substance Use Disorder Treatment: A Narrative Review. ALPHA PSYCHIATRY 2024; 25:206-211. [PMID: 38798813 PMCID: PMC11117434 DOI: 10.5152/alphapsychiatry.2024.241522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/06/2024] [Indexed: 05/29/2024]
Abstract
Substance use disorder (SUD) continues to pose a significant global health challenge, necessitating innovative and effective therapeutic interventions. Ketamine, traditionally recognized for its anesthetic properties, has emerged as a novel and promising avenue for the treatment of SUD. This narrative review critically examines the current body of literature surrounding the use of ketamine in various forms and settings for individuals grappling with substance abuse. The review explores the neurobiological underpinnings of ketamine's potential therapeutic effects in SUD, shedding light on its impact on glutamatergic neurotransmission, neuroplasticity, and reward pathways. Special attention is given to the psychotropic and dissociative properties of ketamine, exploring their implications for both therapeutic outcomes and patient experience. Ultimately, this review aims to provide a comprehensive overview of the current state of knowledge regarding ketamine's role in the treatment of SUD, emphasizing the need for further research and clinical exploration. As we navigate the complex terrain of addiction medicine, understanding the nuances of ketamine's potential in SUD holds promise for the development of more effective and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Anna Famuła
- Department of Psychiatry, Medical University of Silesia, Faculty of Medical Sciences, Zabrze, Poland
- Intravenous Ketamine Treatment Laboratory, Multispecialty County Hospital, Tarnowskie Góry, Poland
| | - Jakub Radoszewski
- Department and Clinical Division of Psychiatry and Psychotherapy of Developmental Age, Medical University of Silesia, Faculty of Medical Sciences, Katowice, Poland
| | - Tomasz Czerwiec
- Department and Clinical Division of Psychiatry and Psychotherapy of Developmental Age, Medical University of Silesia, Faculty of Medical Sciences, Katowice, Poland
| | - Jarosław Sobiś
- Department of Psychiatry, Medical University of Silesia, Faculty of Medical Sciences, Zabrze, Poland
| | - Gniewko Więckiewicz
- Department of Psychiatry, Medical University of Silesia, Faculty of Medical Sciences, Zabrze, Poland
- Intravenous Ketamine Treatment Laboratory, Multispecialty County Hospital, Tarnowskie Góry, Poland
| |
Collapse
|
22
|
Ledesma-Corvi S, Jornet-Plaza J, Gálvez-Melero L, García-Fuster MJ. Novel rapid treatment options for adolescent depression. Pharmacol Res 2024; 201:107085. [PMID: 38309382 DOI: 10.1016/j.phrs.2024.107085] [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: 08/01/2023] [Revised: 12/13/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
There is an urgent need for novel fast-acting antidepressants for adolescent treatment-resistant depression and/or suicidal risk, since the selective serotonin reuptake inhibitors that are clinically approved for that age (i.e., fluoxetine or escitalopram) take weeks to work. In this context, one of the main research lines of our group is to characterize at the preclinical level novel approaches for rapid-acting antidepressants for adolescence. The present review summarizes the potential use in adolescence of non-pharmacological options, such as neuromodulators (electroconvulsive therapy and other innovative types of brain stimulation), as well as pharmacological options, including consciousness-altering drugs (mainly ketamine but also classical psychedelics) and cannabinoids (i.e., cannabidiol), with promising fast-acting responses. Following a brief analytical explanation of adolescent depression, we present a general introduction for each therapeutical approach together with the clinical evidence supporting its potential beneficial use in adolescence (mainly extrapolated from prior successful examples for adults), to then report recent and/or ongoing preclinical studies that will aid in improving the inclusion of these therapies in the clinic, by considering potential sex-, age-, and dose-related differences, and/or other factors that might affect efficacy or long-term safety. Finally, we conclude the review by providing future avenues to maximize treatment response, including the need for more clinical studies and the importance of designing and/or testing novel treatment options that are safe and fast-acting for adolescent depression.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jordi Jornet-Plaza
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Laura Gálvez-Melero
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain; Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
23
|
Aicher HD, Mueller MJ, Dornbierer DA, Suay D, Elsner C, Wicki I, Meling D, Caflisch L, Hempe A, Steinhart C, Mueller J, Von Rotz R, Kleim B, Scheidegger M. Potential therapeutic effects of an ayahuasca-inspired N,N-DMT and harmine formulation: a controlled trial in healthy subjects. Front Psychiatry 2024; 14:1302559. [PMID: 38264636 PMCID: PMC10804806 DOI: 10.3389/fpsyt.2023.1302559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Background There is growing scientific evidence for the therapeutic benefits of the Amazonian plant-based psychedelic "ayahuasca" for neuropsychiatric disorders such as depression and anxiety. However, there are certain challenges when incorporating botanical ayahuasca into biomedical research and clinical therapy environments. Formulations inspired by ayahuasca, which contain specific and standardized active components, are a potential remedy. Methods We investigated subjective acute and persisting effects of a novel formulation containing the reversible monoamine oxidase inhibitor harmine (orodispersible tablet containing 100 mg MAO-I) and N,N-dimethyltryptamine (incremental intranasal dosing of up to 100 mg DMT), compared with two other conditions, namely harmine alone and placebo, in a crossover RCT in 31 healthy male subjects. Results DMT + harmine, but not harmine alone, induced a psychedelic experience assessed with the 5D-ASC rating scale [global score: F(2,60) = 80.21, p < 0.001] and acute experience sampling items over time, characterized by psychological insights [PIQ, F(2,58.5) = 28.514, p < 0.001], emotional breakthroughs [EBI, F(2,60) = 26.509, p < 0.001], and low scores on the challenging experience questionnaire [CEQ, F(2,60) = 12.84, p < 0.001]. Participants attributed personal and spiritual significance to the experience (GSR) with mainly positive persisting effects (PEQ) at 1- and 4-months follow-up. Acute drug effects correlated positively with persisting effects. We found no changes in trait measures of personality, psychological flexibility, or general well-being, and no increases in psychopathology (SCL-90-R) were reported. Discussion and Conclusion Our results suggest that the experience induced by the standardized DMT + harmine formulation induces a phenomenologically rich psychedelic experience, demonstrates good psychological safety and tolerability, is well tolerated, and induces beneficial psychological processes that could possibly support psychotherapy. Further studies are required to investigate the psychotherapeutic potential in patients.
Collapse
Affiliation(s)
- Helena D. Aicher
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Psychology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
| | - Michael J. Mueller
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
- Department of Health Science and Technology, ETH, Zurich, Switzerland
| | - Dario A. Dornbierer
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Department of Forensic Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Dila Suay
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- MoMiLab, IMT School for Advanced Studies Luca, Lucca, Italy
| | - Claudius Elsner
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ilhui Wicki
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniel Meling
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Psychosomatic Medicine and Psychotherapy, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Luzia Caflisch
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alexandra Hempe
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Biopsychology, Department of Psychology, TUD Dresden University of Technology, Dresden, Germany
| | - Camilla Steinhart
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jovin Mueller
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Robin Von Rotz
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Birgit Kleim
- Department of Psychology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
- Experimental Psychopathology and Psychotherapy, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Milan Scheidegger
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
| |
Collapse
|
24
|
Jóźwiak-Bębenista M, Sokołowska P, Wiktorowska-Owczarek A, Kowalczyk E, Sienkiewicz M. Ketamine - A New Antidepressant Drug with Anti-Inflammatory Properties. J Pharmacol Exp Ther 2024; 388:134-144. [PMID: 37977808 DOI: 10.1124/jpet.123.001823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/04/2023] [Accepted: 10/04/2023] [Indexed: 11/19/2023] Open
Abstract
Ketamine is a new, potent and rapid-acting antidepressant approved for therapy of treatment-resistant depression, which has a different mechanism of action than currently-available antidepressant therapies. It owes its uniquely potent antidepressant properties to a complex mechanism of action, which currently remains unclear. However, it is thought that it acts by modulating the functioning of the glutamatergic system, which plays an important role in the process of neuroplasticity associated with depression. However, preclinical and clinical studies have also found ketamine to reduce inflammation, either directly or indirectly (by activating neuroprotective branches of the kynurenine pathway), among patients exhibiting higher levels of inflammation. Inflammation and immune system activation are believed to play key roles in the development and course of depression. Therefore, the present work examines the role of the antidepressant effect of ketamine and its anti-inflammatory properties in the treatment of depression. SIGNIFICANCE STATEMENT: The present work examines the relationship between the antidepressant effect of ketamine and its anti-inflammatory properties, and the resulting benefits in treatment-resistant depression (TRD). The antidepressant mechanism of ketamine remains unclear, and there is an urgent need to develop new therapeutic strategies for treatment of depression, particularly TRD.
Collapse
Affiliation(s)
- Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Paulina Sokołowska
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Anna Wiktorowska-Owczarek
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmacology and Toxicology (M.J.-B., P.S., A.W.-O., E.K.) and Department of Pharmaceutical Microbiology and Microbiological Diagnostics (M.S.), Medical University of Lodz, Lodz, Poland
| |
Collapse
|
25
|
Burback L, Brémault-Phillips S, Nijdam MJ, McFarlane A, Vermetten E. Treatment of Posttraumatic Stress Disorder: A State-of-the-art Review. Curr Neuropharmacol 2024; 22:557-635. [PMID: 37132142 PMCID: PMC10845104 DOI: 10.2174/1570159x21666230428091433] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 05/04/2023] Open
Abstract
This narrative state-of-the-art review paper describes the progress in the understanding and treatment of Posttraumatic Stress Disorder (PTSD). Over the last four decades, the scientific landscape has matured, with many interdisciplinary contributions to understanding its diagnosis, etiology, and epidemiology. Advances in genetics, neurobiology, stress pathophysiology, and brain imaging have made it apparent that chronic PTSD is a systemic disorder with high allostatic load. The current state of PTSD treatment includes a wide variety of pharmacological and psychotherapeutic approaches, of which many are evidence-based. However, the myriad challenges inherent in the disorder, such as individual and systemic barriers to good treatment outcome, comorbidity, emotional dysregulation, suicidality, dissociation, substance use, and trauma-related guilt and shame, often render treatment response suboptimal. These challenges are discussed as drivers for emerging novel treatment approaches, including early interventions in the Golden Hours, pharmacological and psychotherapeutic interventions, medication augmentation interventions, the use of psychedelics, as well as interventions targeting the brain and nervous system. All of this aims to improve symptom relief and clinical outcomes. Finally, a phase orientation to treatment is recognized as a tool to strategize treatment of the disorder, and position interventions in step with the progression of the pathophysiology. Revisions to guidelines and systems of care will be needed to incorporate innovative treatments as evidence emerges and they become mainstream. This generation is well-positioned to address the devastating and often chronic disabling impact of traumatic stress events through holistic, cutting-edge clinical efforts and interdisciplinary research.
Collapse
Affiliation(s)
- Lisa Burback
- Department of Psychiatry, University of Alberta, Edmonton, Canada
| | | | - Mirjam J. Nijdam
- ARQ National Psychotrauma Center, Diemen, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Eric Vermetten
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Department of Psychiatry, New York University Grossman School of Medicine, New York, USA
| |
Collapse
|
26
|
Johnston JN, Kadriu B, Kraus C, Henter ID, Zarate CA. Ketamine in neuropsychiatric disorders: an update. Neuropsychopharmacology 2024; 49:23-40. [PMID: 37340091 PMCID: PMC10700638 DOI: 10.1038/s41386-023-01632-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/08/2023] [Accepted: 06/01/2023] [Indexed: 06/22/2023]
Abstract
The discovery of ketamine as a rapid-acting antidepressant led to a new era in the development of neuropsychiatric therapeutics, one characterized by an antidepressant response that occurred within hours or days rather than weeks or months. Considerable clinical research supports the use of-or further research with-subanesthetic-dose ketamine and its (S)-enantiomer esketamine in multiple neuropsychiatric disorders including depression, bipolar disorder, anxiety spectrum disorders, substance use disorders, and eating disorders, as well as for the management of chronic pain. In addition, ketamine often effectively targets symptom domains associated with multiple disorders, such as anxiety, anhedonia, and suicidal ideation. This manuscript: 1) reviews the literature on the pharmacology and hypothesized mechanisms of subanesthetic-dose ketamine in clinical research; 2) describes similarities and differences in the mechanism of action and antidepressant efficacy between racemic ketamine, its (S) and (R) enantiomers, and its hydroxynorketamine (HNK) metabolite; 3) discusses the day-to-day use of ketamine in the clinical setting; 4) provides an overview of ketamine use in other psychiatric disorders and depression-related comorbidities (e.g., suicidal ideation); and 5) provides insights into the mechanisms of ketamine and therapeutic response gleaned from the study of other novel therapeutics and neuroimaging modalities.
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Translational and Experimental Medicine, Neuroscience at Jazz Pharmaceuticals, San Diego, CA, USA
| | - Christoph Kraus
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Ioline D Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Maddock JE, Johnson SS. Spending Time in Nature: The Overlooked Health Behavior. Am J Health Promot 2024; 38:124-148. [PMID: 38126319 DOI: 10.1177/08901171231210806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Affiliation(s)
- Jay E Maddock
- Department of Environmental and Occupational Health, School of Public Health, Texas A&M University, Center for Health and Nature, College Station, TX, USA
| | | |
Collapse
|
28
|
Su WJ, Hu T, Jiang CL. Cool the Inflamed Brain: A Novel Anti-inflammatory Strategy for the Treatment of Major Depressive Disorder. Curr Neuropharmacol 2024; 22:810-842. [PMID: 37559243 PMCID: PMC10845090 DOI: 10.2174/1570159x21666230809112028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/21/2023] [Accepted: 02/23/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Abundant evidence suggests that inflammatory cytokines contribute to the symptoms of major depressive disorder (MDD) by altering neurotransmission, neuroplasticity, and neuroendocrine processes. Given the unsatisfactory response and remission of monoaminergic antidepressants, anti-inflammatory therapy is proposed as a feasible way to augment the antidepressant effect. Recently, there have been emerging studies investigating the efficiency and efficacy of anti-inflammatory agents in the treatment of MDD and depressive symptoms comorbid with somatic diseases. METHODS In this narrative review, prospective clinical trials focusing on anti-inflammatory treatment for depression have been comprehensively searched and screened. Based on the included studies, we summarize the rationale for the anti-inflammatory therapy of depression and discuss the utilities and confusions regarding the anti-inflammatory strategy for MDD. RESULTS This review included over 45 eligible trials. For ease of discussion, we have grouped them into six categories based on their mechanism of action, and added some other anti-inflammatory modalities, including Chinese herbal medicine and non-drug therapy. Pooled results suggest that anti-inflammatory therapy is effective in improving depressive symptoms, whether used as monotherapy or add-on therapy. However, there remain confusions in the application of anti-inflammatory therapy for MDD. CONCLUSION Based on current clinical evidence, anti-inflammatory therapy is a promisingly effective treatment for depression. This study proposes a novel strategy for clinical diagnosis, disease classification, personalized treatment, and prognostic prediction of depression. Inflammatory biomarkers are recommended to be assessed at the first admission of MDD patients, and anti-inflammatory therapy are recommended to be included in the clinical practice guidelines for diagnosis and treatment. Those patients with high levels of baseline inflammation (e.g., CRP > 3 mg/L) may benefit from adjunctive anti-inflammatory therapy.
Collapse
Affiliation(s)
- Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, 200433, China
| | - Ting Hu
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, 200433, China
| | - Chun-Lei Jiang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
29
|
Zaretsky TG, Jagodnik KM, Barsic R, Antonio JH, Bonanno PA, MacLeod C, Pierce C, Carney H, Morrison MT, Saylor C, Danias G, Lepow L, Yehuda R. The Psychedelic Future of Post-Traumatic Stress Disorder Treatment. Curr Neuropharmacol 2024; 22:636-735. [PMID: 38284341 PMCID: PMC10845102 DOI: 10.2174/1570159x22666231027111147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 01/30/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a mental health condition that can occur following exposure to a traumatic experience. An estimated 12 million U.S. adults are presently affected by this disorder. Current treatments include psychological therapies (e.g., exposure-based interventions) and pharmacological treatments (e.g., selective serotonin reuptake inhibitors (SSRIs)). However, a significant proportion of patients receiving standard-of-care therapies for PTSD remain symptomatic, and new approaches for this and other trauma-related mental health conditions are greatly needed. Psychedelic compounds that alter cognition, perception, and mood are currently being examined for their efficacy in treating PTSD despite their current status as Drug Enforcement Administration (DEA)- scheduled substances. Initial clinical trials have demonstrated the potential value of psychedelicassisted therapy to treat PTSD and other psychiatric disorders. In this comprehensive review, we summarize the state of the science of PTSD clinical care, including current treatments and their shortcomings. We review clinical studies of psychedelic interventions to treat PTSD, trauma-related disorders, and common comorbidities. The classic psychedelics psilocybin, lysergic acid diethylamide (LSD), and N,N-dimethyltryptamine (DMT) and DMT-containing ayahuasca, as well as the entactogen 3,4-methylenedioxymethamphetamine (MDMA) and the dissociative anesthetic ketamine, are reviewed. For each drug, we present the history of use, psychological and somatic effects, pharmacology, and safety profile. The rationale and proposed mechanisms for use in treating PTSD and traumarelated disorders are discussed. This review concludes with an in-depth consideration of future directions for the psychiatric applications of psychedelics to maximize therapeutic benefit and minimize risk in individuals and communities impacted by trauma-related conditions.
Collapse
Affiliation(s)
- Tamar Glatman Zaretsky
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kathleen M. Jagodnik
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Barsic
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josimar Hernandez Antonio
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip A. Bonanno
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn MacLeod
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charlotte Pierce
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hunter Carney
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Morgan T. Morrison
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles Saylor
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George Danias
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Lepow
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Yehuda
- James J. Peters Veterans Affairs Medical Center, New York, NY, USA
- The Center for Psychedelic Psychotherapy and Trauma Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
30
|
Witt CE, Mena S, Holmes J, Hersey M, Buchanan AM, Parke B, Saylor R, Honan LE, Berger SN, Lumbreras S, Nijhout FH, Reed MC, Best J, Fadel J, Schloss P, Lau T, Hashemi P. Serotonin is a common thread linking different classes of antidepressants. Cell Chem Biol 2023; 30:1557-1570.e6. [PMID: 37992715 DOI: 10.1016/j.chembiol.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 09/07/2023] [Accepted: 10/12/2023] [Indexed: 11/24/2023]
Abstract
Depression pathology remains elusive. The monoamine hypothesis has placed much focus on serotonin, but due to the variable clinical efficacy of monoamine reuptake inhibitors, the community is looking for alternative therapies such as ketamine (neurogenesis theory of antidepressant action). There is evidence that different classes of antidepressants may affect serotonin levels; a notion we test here. We measure hippocampal serotonin in mice with voltammetry and study the effects of acute challenges of escitalopram, fluoxetine, reboxetine, and ketamine. We find that pseudo-equivalent doses of these drugs similarly raise ambient serotonin levels, despite their differing pharmacodynamics because of differences in Uptake 1 and 2, rapid SERT trafficking, and modulation of serotonin by histamine. These antidepressants have different pharmacodynamics but have strikingly similar effects on extracellular serotonin. Our findings suggest that serotonin is a common thread that links clinically effective antidepressants, synergizing different theories of depression (synaptic plasticity, neurogenesis, and the monoamine hypothesis).
Collapse
Affiliation(s)
- Colby E Witt
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Sergio Mena
- Department of Bioengineering, Imperial College London, London, UK
| | - Jordan Holmes
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Melinda Hersey
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA; Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Anna Marie Buchanan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA; Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Brenna Parke
- Department of Bioengineering, Imperial College London, London, UK
| | - Rachel Saylor
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Lauren E Honan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Shane N Berger
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Sara Lumbreras
- Department of Psychiatry and Psychotherapy, Biochemical Laboratory, Central Institute of Mental Health, Medical Faculty, Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Michael C Reed
- Department of Mathematics, Duke University, Durham, NC, USA
| | - Janet Best
- Department of Mathematics, The Ohio State University, Columbus, OH, USA
| | - James Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Patrick Schloss
- Department of Psychiatry and Psychotherapy, Biochemical Laboratory, Central Institute of Mental Health, Medical Faculty, Mannheim, Heidelberg University, Mannheim, Germany
| | - Thorsten Lau
- Department of Psychiatry and Psychotherapy, Biochemical Laboratory, Central Institute of Mental Health, Medical Faculty, Mannheim, Heidelberg University, Mannheim, Germany; Department of Neuroanatomy, Mannheim Centre for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA; Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
31
|
Brandão AAC, Deus DLS, Duarte-Filho LAMS, Menezes PMN, Massaranduba ABR, Silva FS, Ribeiro LAA. Nebulized and intraperitoneal ketamine have equivalent antidepressant-like effect in the forced swim and tail suspension tests in mice. Pharmacol Biochem Behav 2023; 233:173674. [PMID: 37949377 DOI: 10.1016/j.pbb.2023.173674] [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: 05/19/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Major depressive disorder (MDD) is a debilitating illness that affects millions of people worldwide. Currently available antidepressants often take weeks to months to reach their full effect, which leads to an increased risk of suicidal behavior in patients with MMD. Intranasally, esketamine has emerged as an alternative to current antidepressants because of its rapid onset and long-lasting effects in patients with MDD. Animal models are useful for the initial pharmacological screening and for a better understanding of the mechanisms underlying the effects of new drugs with potential against MDD. There is a lack of data on alternative routes of drug administration, either oral or injectable, that can be used in preclinical studies. This study aimed to test whether ketamine has antidepressant-like effects in mice when administered via nebulization using a low-cost apparatus. When mice whose depressive-like behavior was induced by corticosterone were treated with nebulized ketamine at concentrations of 1.3, 2.6, and 5.2 mg/mL, immobility was reduced by 38.6 %, 62.0 %, and 61.1 %, respectively, in the forced swimming test (FST) and 43.6 %, 42.1 %, and 57.9 %, respectively, in the tail suspension test (TST). When depression-like behavior was induced by dexamethasone, nebulization with ketamine reduced immobility by 79.7 %, 49.2 %, and 44.4 % in the FST and 80.9 %, 71.4 %, and 80.4 %, respectively, in the TST. When depression-like behavior was induced by the association between dexamethasone and unpredictable chronic mild stress (UCMS) exposure, immobility was reduced by 26.1 %, 55.3 %, and 19.1 % in FST. Mice treated with nebulized ketamine did not show significant changes in the distance covered or in the time spent moving in the open field test. The efficacy of intraperitoneal and nebulized ketamine is equivalent, which shows that nebulization can be an alternative inexpensive route of drug administration for behavioral studies in rodents.
Collapse
Affiliation(s)
- Aida A C Brandão
- Curso de mestrado em biociências, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Deborah L S Deus
- Curso de graduação em farmácia, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Luiz A M S Duarte-Filho
- UMR CNRS 7266 LIENSs, Département de Biotechnologie, La Rochelle Université, La Rochelle, France
| | - Pedro M N Menezes
- Faculdade Maurício de Nassau (UNINASSAU), Av. Cardoso de Sá, 950, Cidade Universitária, Petrolina, PE CEP: 56328-020, Brazil; Faculdade de Petrolina (FACAPE), Campus Universitário, s/n, Vila Eduardo, Petrolina, PE CEP: 56328-903, Brazil
| | - Ana B R Massaranduba
- Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Fabrício S Silva
- Colegiado de Ciências Farmacêuticas (CFARM), Programa de Pós-graduação em Biociências (PGB), Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Luciano A A Ribeiro
- Colegiado de Ciências Farmacêuticas (CFARM), Programa de Pós-graduação em Biociências (PGB), Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil.
| |
Collapse
|
32
|
Gomez‐Mancilla B, Levy JA, Ganesan S, Faller T, Issachar G, Peremen Z, Laufer O, Shani‐Hershkovich R, Biliouris K, Walker E, Healy MP, Sverdlov O, Desai S, Ghaemi SN, Cha J, Shanker YG. MIJ821 (onfasprodil) in healthy volunteers: First-in-human, randomized, placebo-controlled study (single ascending dose and repeated intravenous dose). Clin Transl Sci 2023; 16:2236-2252. [PMID: 37817426 PMCID: PMC10651655 DOI: 10.1111/cts.13623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 10/12/2023] Open
Abstract
This single-center study administered MIJ821 (onfasprodil) as an intravenous infusion to healthy volunteers and included two parts: a single ascending dose study (Part 1) and a repeated intravenous dose study (Part 2). Primary objective was to evaluate the safety and tolerability of single ascending intravenous doses infused over a 40-min period and of two repeated doses (1 week apart) of MIJ821 in healthy volunteers. Secondary objectives were to assess the pharmacokinetics of MIJ821 after intravenous infusion in Part 1 and Part 2 of the study. Overall, 43 subjects in Part 1 and 12 subjects in Part 2 were randomized in the study. Median age in Part 1 and Part 2 was 45.0 and 43.5 years, respectively, with the majority being Caucasian (Part 1: 84%; Part 2: 92%). 19 subjects (44.2%) in Part 1 and 8 subjects (66.7%) in Part 2 experienced at least one adverse event (AE). Following single dose in Part 1 and Part 2, the AUCinf values of MIJ821 increased in a dose-proportional manner across the dose range 0.016-0.48 mg/kg and the Cmax values in a slight overproportional manner across the dose range 0.048-0.48 mg/kg. At the highest dose of 0.48 mg/kg, the geometric mean AUCinf was 708 h ng/mL and the geometric mean Cmax was 462 ng/mL. Inspection of 1-h post-dose resting electroencephalography activity across cohorts showed a relationship to administered dose, providing exploratory evidence of distal target engagement. In conclusion, MIJ821 showed a good safety and tolerability profile in healthy volunteers. Dissociative AEs were mild, transient, and dose-dependent.
Collapse
Affiliation(s)
- Baltazar Gomez‐Mancilla
- Novartis Institutes for BioMedical ResearchNovartis Pharma AGBaselSwitzerland
- McGill UniversityMontrealQuébecCanada
| | | | | | - Thomas Faller
- Novartis Institutes for BioMedical ResearchNovartis Pharma AGBaselSwitzerland
| | | | | | | | | | - Kostas Biliouris
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | - Ela Walker
- Novartis Institutes for BioMedical ResearchNovartis Pharma AGBaselSwitzerland
| | - Mark P. Healy
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | | | - Sachin Desai
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| | - S. Nassir Ghaemi
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
- Tufts Medical Center, Tufts UniversityBostonMassachusettsUSA
| | - Jang‐Ho Cha
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | - Y. Gopi Shanker
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
- Beam TherapeuticsCambridgeMassachusettsUSA
| |
Collapse
|
33
|
Agüera ADR, Cándido C, Donaire R, Papini MR, Torres C. Ketamine retards recovery from reward downshift and supports conditioned taste aversion. Pharmacol Biochem Behav 2023; 233:173671. [PMID: 39492495 DOI: 10.1016/j.pbb.2023.173671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Ketamine is a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist with antidepressant, anxiolytic, and memory effects in clinical and preclinical studies. The present studies investigated the behavioral effects of ketamine in animals exposed to a consummatory successive negative contrast (cSNC) task involving unexpected reward downshift, negative emotion (frustration), and aversive memory. Food-restricted male rats had 5-min access to 32 % sucrose in each of 10 preshift sessions followed by 4 % sucrose in 4 postshift sessions. Unshifted controls had access to 4 % sucrose during all 14 sessions. Ketamine (10 mg/kg, ip) was injected 30 min before sessions 11 and 12 (Experiment 1) or immediately after session 11 (Experiment 3). The results showed that both pre- and postdownshift session injection of ketamine increased consummatory suppression, as Group 32/Ket exhibited lower sucrose intake than Groups 32/Sal, 4/Ket, and 4/Sal. These effects extended beyond the day(s) of injection. Experiments 2 and 4 showed that the same dose, route of administration, and time of injection induced significant conditioned taste aversion to 4 % sucrose, in the absence of reward downshift. These data suggest that ketamine induces an aversive state that may summate with frustration induced by reward downshift in the cSNC task and also support a conditioned taste aversion to 4 % sucrose in the absence of reward downshift. Implications for these and other experiments involving pre- and postsession administration of ketamine are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Carmen Torres
- Department of Psychology, University of Jaén, Spain.
| |
Collapse
|
34
|
Dębowska W, Więdłocha M, Dębowska M, Kownacka Z, Marcinowicz P, Szulc A. Transcranial magnetic stimulation and ketamine: implications for combined treatment in depression. Front Neurosci 2023; 17:1267647. [PMID: 37954877 PMCID: PMC10637948 DOI: 10.3389/fnins.2023.1267647] [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: 07/27/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Drug-resistant mental disorders, particularly treatment-resistant depression, pose a significant medical and social problem. To address this challenge, modern psychiatry is constantly exploring the use of novel treatment methods, including biological treatments, such as transcranial magnetic stimulation (TMS), and novel rapid-acting antidepressants, such as ketamine. While both TMS and ketamine demonstrate high effectiveness in reducing the severity of depressive symptoms, some patients still do not achieve the desired improvement. Recent literature suggests that combining these two methods may yield even stronger and longer-lasting results. This review aims to consolidate knowledge in this area and elucidate the potential mechanisms of action underlying the increased efficacy of combined treatment, which would provide a foundation for the development and optimization of future treatment protocols.
Collapse
Affiliation(s)
- Weronika Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Więdłocha
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- KeyClinic, Warsaw, Poland
| | - Marta Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Kownacka
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Marcinowicz
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- KeyClinic, Warsaw, Poland
| | - Agata Szulc
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- MindHealth, Warsaw, Poland
| |
Collapse
|
35
|
Akbar D, Rhee TG, Ceban F, Ho R, Teopiz KM, Cao B, Subramaniapillai M, Kwan ATH, Rosenblat JD, McIntyre RS. Dextromethorphan-Bupropion for the Treatment of Depression: A Systematic Review of Efficacy and Safety in Clinical Trials. CNS Drugs 2023; 37:867-881. [PMID: 37792265 DOI: 10.1007/s40263-023-01032-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND A significant proportion of adults with major depressive disorder (MDD) do not respond to treatments which are currently used in clinical practice such as first-generation monoamine-based antidepressants. OBJECTIVES The objective of this systematic review was to assess the efficacy, safety, and mechanisms of action of AXS-05, a combination of the NMDA-receptor antagonist dextromethorphan with bupropion, in adults with MDD. METHODS We searched PubMed, Embase, Google Scholar, and ClinicalTrials.gov for current studies reporting on efficacy and/or safety of AXS-05 in patients with MDD. The search terms included: "AXS-05" OR "dextromethorphan and bupropion" AND "depression". Studies from database inception to January 2023 were evaluated. Risk of bias was assessed using the Cochrane Risk of Bias tool. RESULTS The search yielded 54 studies of which 5 were included. All studies had low risk of bias. Depression severity, measured with the Montgomery-Åsberg Depression Rating Scale (MADRS) significantly decreased as early as 1-week post-treatment from baseline when compared to a placebo-controlled group (LS mean difference 2.2; 95% CI 0.6-3.9; p = 0.007) and at 2 weeks compared to an active control group (LS mean difference 4.7; 95% CI 0.6-8.8; p = 0.024). Treatment efficacy could be maintained for up to 12 months with mean MADRS score reduction of 23 points from baseline. Clinical remission and response rates also improved at week 1 and were maintained for 12 months. The treatment was well-tolerated, with some transient adverse events reported. CONCLUSION Current evidence suggests that the combination of dextromethorphan and bupropion is a well-tolerated, rapid-acting treatment option for adults with MDD. Initial success with AXS-05 supports the mechanistic role of glutamatergeric and sigma 1 signaling in the pathophysiology of MDD.
Collapse
Affiliation(s)
- Dania Akbar
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Taeho Greg Rhee
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- VA New England Mental Illness, Research, Education and Clinical Center (MIRECC), VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Public Health Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Felicia Ceban
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Brain and Cognition Discovery Foundation (BCDF), 77 Bloor St W Suite 617, Toronto, ON, M5S 1M2, Canada
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Roger Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Kayla M Teopiz
- Brain and Cognition Discovery Foundation (BCDF), 77 Bloor St W Suite 617, Toronto, ON, M5S 1M2, Canada
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, 400715, People's Republic of China
| | - Mehala Subramaniapillai
- Brain and Cognition Discovery Foundation (BCDF), 77 Bloor St W Suite 617, Toronto, ON, M5S 1M2, Canada
| | - Angela T H Kwan
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Brain and Cognition Discovery Foundation (BCDF), 77 Bloor St W Suite 617, Toronto, ON, M5S 1M2, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Joshua D Rosenblat
- Mood Disorder Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Brain and Cognition Discovery Foundation (BCDF), 77 Bloor St W Suite 617, Toronto, ON, M5S 1M2, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
36
|
Zelada MI, Garrido V, Liberona A, Jones N, Zúñiga K, Silva H, Nieto RR. Brain-Derived Neurotrophic Factor (BDNF) as a Predictor of Treatment Response in Major Depressive Disorder (MDD): A Systematic Review. Int J Mol Sci 2023; 24:14810. [PMID: 37834258 PMCID: PMC10572866 DOI: 10.3390/ijms241914810] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/16/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has been studied as a biomarker of major depressive disorder (MDD). Besides diagnostic biomarkers, clinically useful biomarkers can inform response to treatment. We aimed to review all studies that sought to relate BDNF baseline levels, or BDNF polymorphisms, with response to treatment in MDD. In order to achieve this, we performed a systematic review of studies that explored the relation of BDNF with both pharmacological and non-pharmacological treatment. Finally, we reviewed the evidence that relates peripheral levels of BDNF and BDNF polymorphisms with the development and management of treatment-resistant depression.
Collapse
Affiliation(s)
- Mario Ignacio Zelada
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Verónica Garrido
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Andrés Liberona
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Natalia Jones
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Karen Zúñiga
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Hernán Silva
- Clínica Psiquiátrica Universitaria, Hospital Clínico de la Universidad de Chile, Universidad de Chile, Santiago 8380453, Chile
- Departamento de Psiquiatría y Salud Mental Norte, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Rodrigo R. Nieto
- Clínica Psiquiátrica Universitaria, Hospital Clínico de la Universidad de Chile, Universidad de Chile, Santiago 8380453, Chile
- Departamento de Psiquiatría y Salud Mental Norte, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Departamento de Neurociencias, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| |
Collapse
|
37
|
Spencer HF, Boese M, Berman RY, Radford KD, Choi KH. Effects of a Subanesthetic Ketamine Infusion on Inflammatory and Behavioral Outcomes after Closed Head Injury in Rats. Bioengineering (Basel) 2023; 10:941. [PMID: 37627826 PMCID: PMC10452037 DOI: 10.3390/bioengineering10080941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Traumatic brain injury (TBI) affects millions of people annually, and most cases are classified as mild TBI (mTBI). Ketamine is a potent trauma analgesic and anesthetic with anti-inflammatory properties. However, ketamine's effects on post-mTBI outcomes are not well characterized. For the current study, we used the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA), which replicates the biomechanics of a closed-head impact with resulting free head movement. Adult male Sprague-Dawley rats sustained a single-session, repeated-impacts CHIMERA injury. An hour after the injury, rats received an intravenous ketamine infusion (0, 10, or 20 mg/kg, 2 h period), during which locomotor activity was monitored. Catheter blood samples were collected at 1, 3, 5, and 24 h after the CHIMERA injury for plasma cytokine assays. Behavioral assays were conducted on post-injury days (PID) 1 to 4 and included rotarod, locomotor activity, acoustic startle reflex (ASR), and pre-pulse inhibition (PPI). Brain tissue samples were collected at PID 4 and processed for GFAP (astrocytes), Iba-1 (microglia), and silver staining (axonal injury). Ketamine dose-dependently altered locomotor activity during the infusion and reduced KC/GRO, TNF-α, and IL-1β levels after the infusion. CHIMERA produced a delayed deficit in rotarod performance (PID 3) and significant axonal damage in the optic tract (PID 4), without significant changes in other behavioral or histological measures. Notably, subanesthetic doses of intravenous ketamine infusion after mTBI did not produce adverse effects on behavioral outcomes in PID 1-4 or neuroinflammation on PID 4. A further study is warranted to thoroughly investigate beneficial effects of IV ketamine on mTBI given multi-modal properties of ketamine in traumatic injury and stress.
Collapse
Affiliation(s)
- Haley F. Spencer
- Program in Neuroscience, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA;
- Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA;
| | - Martin Boese
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Rina Y. Berman
- Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA;
| | - Kennett D. Radford
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
| | - Kwang H. Choi
- Program in Neuroscience, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA;
- Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA;
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA; (M.B.); (K.D.R.)
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Services University, 4301 Jones Bridge Rd, Bethesda, MD 20814, USA
| |
Collapse
|
38
|
Ryan K, Hosanagar A. Ketamine Use in Child and Adolescent Psychiatry: Emerging Data in Treatment-Resistant Depression, Insights from Adults, and Future Directions. Curr Psychiatry Rep 2023; 25:337-344. [PMID: 37389787 DOI: 10.1007/s11920-023-01432-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 07/01/2023]
Abstract
PURPOSE OF REVIEW The following review will explore ketamine's antidepressant and antisuicidal properties in adults, review of what is known about ketamine's safety in children, and summarize the limited information we have on ketamine's role in treating depression and suicidal ideation in adolescents with depression. Future directions for ketamine's role in child psychiatry based on animal and adult studies will also be explored. RECENT FINDINGS Over the past 20 years, ketamine has emerged as a novel treatment for depression and suicidal ideation in adults. In recent years, these studies have been extended to adolescents. In 2021, the first placebo-controlled trial examining ketamine's antidepressant potential in adolescents was performed, demonstrating superior efficacy over midazolam. Initial studies suggest that ketamine functions as a rapidly acting antidepressant in adolescents. Case reports suggest that ketamine may also reduce suicidal ideation in this population. However, existing studies are small, and more research is needed to solidify these findings and inform clinical practice.
Collapse
Affiliation(s)
- Kaitlyn Ryan
- Department of Psychiatry, Adolescent Partial Hospitalization Program, Trinity Health Ann Arbor Hospital, Ann Arbor, MI, USA.
| | - Avinash Hosanagar
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, USA
- Mental Health Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
39
|
Charlton CE, Karvelis P, McIntyre RS, Diaconescu AO. Suicide prevention and ketamine: insights from computational modeling. Front Psychiatry 2023; 14:1214018. [PMID: 37457775 PMCID: PMC10342546 DOI: 10.3389/fpsyt.2023.1214018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Suicide is a pressing public health issue, with over 700,000 individuals dying each year. Ketamine has emerged as a promising treatment for suicidal thoughts and behaviors (STBs), yet the complex mechanisms underlying ketamine's anti-suicidal effect are not fully understood. Computational psychiatry provides a promising framework for exploring the dynamic interactions underlying suicidality and ketamine's therapeutic action, offering insight into potential biomarkers, treatment targets, and the underlying mechanisms of both. This paper provides an overview of current computational theories of suicidality and ketamine's mechanism of action, and discusses various computational modeling approaches that attempt to explain ketamine's anti-suicidal effect. More specifically, the therapeutic potential of ketamine is explored in the context of the mismatch negativity and the predictive coding framework, by considering neurocircuits involved in learning and decision-making, and investigating altered connectivity strengths and receptor densities targeted by ketamine. Theory-driven computational models offer a promising approach to integrate existing knowledge of suicidality and ketamine, and for the extraction of model-derived mechanistic parameters that can be used to identify patient subgroups and personalized treatment approaches. Future computational studies on ketamine's mechanism of action should optimize task design and modeling approaches to ensure parameter reliability, and external factors such as set and setting, as well as psychedelic-assisted therapy should be evaluated for their additional therapeutic value.
Collapse
Affiliation(s)
- Colleen E. Charlton
- Krembil Center for Neuroinformatics, Center for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Povilas Karvelis
- Krembil Center for Neuroinformatics, Center for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Roger S. McIntyre
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Andreea O. Diaconescu
- Krembil Center for Neuroinformatics, Center for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Cutler AJ, Mattingly GW, Maletic V. Understanding the mechanism of action and clinical effects of neuroactive steroids and GABAergic compounds in major depressive disorder. Transl Psychiatry 2023; 13:228. [PMID: 37365161 PMCID: PMC10293235 DOI: 10.1038/s41398-023-02514-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/12/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
The pathophysiology of major depressive disorder (MDD) is thought to result from impaired connectivity between key brain networks. Gamma-aminobutyric acid (GABA) is the key inhibitory neurotransmitter in the brain, working primarily via GABAA receptors, with an important role in virtually all physiologic functions in the brain. Some neuroactive steroids (NASs) are positive allosteric modulators (PAMs) of GABAA receptors and potentiate phasic and tonic inhibitory responses via activation of synaptic and extrasynaptic GABAA receptors, respectively. This review first discusses preclinical and clinical data that support the association of depression with diverse defects in the GABAergic system of neurotransmission. Decreased levels of GABA and NASs have been observed in adults with depression compared with healthy controls, while treatment with antidepressants normalized the altered levels of GABA and NASs. Second, as there has been intense interest in treatment approaches for depression that target dysregulated GABAergic neurotransmission, we discuss NASs approved or currently in clinical development for the treatment of depression. Brexanolone, an intravenous NAS and a GABAA receptor PAM, is approved by the U.S. Food and Drug Administration for the treatment of postpartum depression (PPD) in patients 15 years and older. Other NASs include zuranolone, an investigational oral GABAA receptor PAM, and PH10, which acts on nasal chemosensory receptors; clinical data to date have shown improvement in depressive symptoms with these investigational NASs in adults with MDD or PPD. Finally, the review discusses how NAS GABAA receptor PAMs may potentially address the unmet need for novel and effective treatments with rapid and sustained antidepressant effects in patients with MDD.
Collapse
|
41
|
Lii TR, Singh V. Ketamine for Complex Regional Pain Syndrome: A Narrative Review Highlighting Dosing Practices and Treatment Response. Anesthesiol Clin 2023; 41:357-369. [PMID: 37245947 PMCID: PMC10688501 DOI: 10.1016/j.anclin.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
This is a narrative review of intravenous ketamine infusions for the treatment of complex regional pain syndrome (CRPS). It briefly covers the definition of CRPS, its epidemiology, and other treatments before introducing ketamine as the article's focus. A summary of ketamine's evidence base and its mechanisms of action is provided. The authors then review ketamine dosages reported in peer-reviewed literature for the treatment of CRPS, and their associated duration of pain relief. The observed response rates to ketamine and predictors of treatment response are also discussed.
Collapse
Affiliation(s)
- Theresa R Lii
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, 450 Broadway Street, MC6343, Redwood City, CA 94063, USA
| | - Vinita Singh
- Department of Anesthesiology, Emory University, 550 Peachtree Street, Emory University Hospital Midtown, Atlanta, GA 30308, USA.
| |
Collapse
|
42
|
Keeler JL, Treasure J, Himmerich H, Brendle M, Moore C, Robison R. Case report: Intramuscular ketamine or intranasal esketamine as a treatment in four patients with major depressive disorder and comorbid anorexia nervosa. Front Psychiatry 2023; 14:1181447. [PMID: 37255674 PMCID: PMC10225569 DOI: 10.3389/fpsyt.2023.1181447] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/18/2023] [Indexed: 06/01/2023] Open
Abstract
Introduction A comorbid diagnosis of a depressive disorder is a negative prognostic factor for individuals with AN, and novel treatments are needed to target depressive symptoms in this population. One emerging promising treatment for depressive disorders is ketamine, although there is less research investigating the use of ketamine for alleviating depression in people with AN. Case report This study reports on four patients with a lifetime diagnosis of AN and a comorbid diagnosis of major depressive disorder who received either intramuscular ketamine (n = 2) or intranasal esketamine (n = 2) treatment from a private psychiatric clinic. Depressive symptomatology (PHQ-9) was measured prior to (es)ketamine administration on every dosing session and adverse effects were recorded during and after dosing. All patients reported a subjective decrease in depression, although only those administered intranasal esketamine showed a reduction in PHQ-9 depression scores over time. Number of doses ranged from 3 to 23. All patients tolerated treatment well and no serious adverse effects emerged, however nausea/vomiting was experienced by one patient on one dosing session. Weight remained stable in all cases, although notably across all patients, weight at the beginning of treatment was within a "healthy" range. Discussion These findings suggest that (es)ketamine may reduce depressive symptoms in people with major depressive disorder and a comorbid diagnosis of AN. Future feasibility and pilot trials are warranted in order to elicit robust data on efficacy, acceptability, safety and tolerability.
Collapse
Affiliation(s)
- Johanna Louise Keeler
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| | - Janet Treasure
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Beckenham, Kent, United Kingdom
| | - Hubertus Himmerich
- Department of Psychological Medicine, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Beckenham, Kent, United Kingdom
| | - Madeline Brendle
- Department of Pharmacotherapy, University of Utah College of Pharmacy, Salt Lake City, UT, United States
- Numinus Wellness, Draper, UT, United States
| | | | - Reid Robison
- Numinus Wellness, Draper, UT, United States
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
43
|
Riccardi A, Guarino M, Serra S, Spampinato MD, Vanni S, Shiffer D, Voza A, Fabbri A, De Iaco F. Narrative Review: Low-Dose Ketamine for Pain Management. J Clin Med 2023; 12:jcm12093256. [PMID: 37176696 PMCID: PMC10179418 DOI: 10.3390/jcm12093256] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Pain is the leading cause of medical consultations and occurs in 50-70% of emergency department visits. To date, several drugs have been used to manage pain. The clinical use of ketamine began in the 1960s and it immediately emerged as a manageable and safe drug for sedation and anesthesia. The analgesic properties of this drug were first reported shortly after its use; however, its psychomimetic effects have limited its use in emergency departments. Owing to the misuse and abuse of opioids in some countries worldwide, ketamine has become a versatile tool for sedation and analgesia. In this narrative review, ketamine's role as an analgesic is discussed, with both known and new applications in various contexts (acute, chronic, and neuropathic pain), along with its strengths and weaknesses, especially in terms of psychomimetic, cardiovascular, and hepatic effects. Moreover, new scientific evidence has been reviewed on the use of additional drugs with ketamine, such as magnesium infusion for improving analgesia and clonidine for treating psychomimetic symptoms. Finally, this narrative review was refined by the experience of the Pain Group of the Italian Society of Emergency Medicine (SIMEU) in treating acute and chronic pain with acute manifestations in Italian Emergency Departments.
Collapse
Affiliation(s)
| | - Mario Guarino
- Emergency Department, Centro Traumatologico Ortopedico, Azienda Ospedaliera di Rilievo Nazionale dei Colli, 80131 Napoli, Italy
| | - Sossio Serra
- Emergency Department, Maurizio Bufalini Hospital, 47522 Cesena, Italy
| | | | - Simone Vanni
- Dipartimento Emergenza e Area Critica, Azienda USL Toscana Centro Struttura Complessa di Medicina d'Urgenza, 50053 Empoli, Italy
| | - Dana Shiffer
- Emergency Department, Humanitas University, Via Rita Levi Montalcini 4, 20089 Milan, Italy
| | - Antonio Voza
- Emergency Department, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Andrea Fabbri
- Emergency Department, AUSL Romagna, Presidio Ospedaliero Morgagni-Pierantoni, 47121 Forlì, Italy
| | - Fabio De Iaco
- Emergency Department, Ospedale Maria Vittoria, 10144 Turin, Italy
| |
Collapse
|
44
|
Tóth A, Sviatkó K, Détári L, Hajnik T. Ketamine affects homeostatic sleep regulation in the absence of the circadian sleep-regulating component in freely moving rats. Pharmacol Biochem Behav 2023; 225:173556. [PMID: 37087059 DOI: 10.1016/j.pbb.2023.173556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
Pharmacological effects of ketamine may affect homeostatic sleep regulation via slow wave related mechanisms. In the present study effects of ketamine applied at anesthetic dose (80 mg/kg) were tested on neocortical electric activity for 24 h in freely moving rats. Ketamine effects were compared to changes during control (saline) injections and after 6 h gentle handling sleep deprivation (SD). As circadian factors may mask drug effects, an illumination protocol consisting of short light-dark cycles was applied. Ketamine application induced a short hypnotic stage with characteristic slow cortical rhythm followed by a long-lasting hyperactive waking resulting pharmacological SD. Coherence analysis indicated an increased level of local synchronization in broad local field potential frequency ranges during hyperactive waking but not during natural- or SD-evoked waking. Both slow wave sleep and rapid eye movement sleep were replaced after the termination of the ketamine effect. Our results show that both ketamine-induced hypnotic state and hyperactive waking can induce homeostatic sleep pressure with comparable intensity as 6 h SD, but ketamine-induced waking was different compared to the SD-evoked one. Both types of waking stages were different compared to spontaneous waking but all three types of wakefulness can engage the homeostatic sleep regulating machinery to generate sleep pressure dissipated by subsequent sleep. Current-source density analysis of the slow waves showed that cortical transmembrane currents were stronger during ketamine-induced hypnotic stage compared to both sleep replacement after SD and ketamine application, but intracortical activation patterns showed only quantitative differences. These findings may hold some translational value for human medical ketamine applications aiming the treatment of depression-associated sleep problems, which can be alleviated by the homeostatic sleep effect of the drug without the need for an intact circadian regulation.
Collapse
Affiliation(s)
- Attila Tóth
- In vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Institute of Biology, Eötvös Loránd University, Hungary.
| | - Katalin Sviatkó
- In vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Institute of Biology, Eötvös Loránd University, Hungary
| | - László Détári
- In vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Institute of Biology, Eötvös Loránd University, Hungary
| | - Tünde Hajnik
- In vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Institute of Biology, Eötvös Loránd University, Hungary
| |
Collapse
|
45
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
46
|
Johnston JN, Kadriu B, Allen J, Gilbert JR, Henter ID, Zarate CA. Ketamine and serotonergic psychedelics: An update on the mechanisms and biosignatures underlying rapid-acting antidepressant treatment. Neuropharmacology 2023; 226:109422. [PMID: 36646310 PMCID: PMC9983360 DOI: 10.1016/j.neuropharm.2023.109422] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
The discovery of ketamine as a rapid-acting antidepressant spurred significant research to understand its underlying mechanisms of action and to identify other novel compounds that may act similarly. Serotonergic psychedelics (SPs) have shown initial promise in treating depression, though the challenge of conducting randomized controlled trials with SPs and the necessity of long-term clinical observation are important limitations. This review summarizes the similarities and differences between the psychoactive effects associated with both ketamine and SPs and the mechanisms of action of these compounds, with a focus on the monoaminergic, glutamatergic, gamma-aminobutyric acid (GABA)-ergic, opioid, and inflammatory systems. Both molecular and neuroimaging aspects are considered. While their main mechanisms of action differ-SPs increase serotonergic signaling while ketamine is a glutamatergic modulator-evidence suggests that the downstream mechanisms of action of both ketamine and SPs include mechanistic target of rapamycin complex 1 (mTORC1) signaling and downstream GABAA receptor activity. The similarities in downstream mechanisms may explain why ketamine, and potentially SPs, exert rapid-acting antidepressant effects. However, research on SPs is still in its infancy compared to the ongoing research that has been conducted with ketamine. For both therapeutics, issues with regulation and proper controls should be addressed before more widespread implementation. This article is part of the Special Issue on "Ketamine and its Metabolites".
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Josh Allen
- The Alfred Centre, Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Ioline D Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
47
|
Qin Y, Guo X, Song W, Liang Z, Wang Y, Feng D, Yang Y, Li M, Gao M. Antidepressant-like effect of CP-101,606: Evidence of mTOR pathway activation. Mol Cell Neurosci 2023; 124:103821. [PMID: 36775184 DOI: 10.1016/j.mcn.2023.103821] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND As a non-competitive N-methyl d-aspartate receptor antagonist, ketamine exerts rapid-onset and long-lasting antidepressant effects on depression, but some side effects limit its use. To identify a safer compound that may provide similar antidepressant effects, here we investigated whether CP-101,606, a selective NR2B receptor inhibitor, provides similar antidepressant effects and explored its underlying mechanisms. METHODS To mimic depressive-like behavior, mice were subjected to chronic unpredictable mild stress (CUMS) for 21 days. Mice were treated with CP-101,606 at 10, 20, and 40 mg/kg doses for 7, 14, and 21 days, respectively, followed by a sucrose preference test (SPT), tail suspension test (TST), and forced swimming test (FST). Western blot analysis was performed on several targets (mTOR, p-mTOR, p70S6K, p-p70S6K, PSD-95, and GluA1), along with immunohistochemistry (GluA1) and immunofluorescence (p-mTOR) assays, using hippocampal tissue. RESULTS CP-101,606 at 20 and 40 mg/kg doses for 7 and 14 days and fluoxetine 10 mg/kg and CP-101606 20 mg/kg for 21 days ameliorated depression-like behaviors in the SPT, TST, and FST. The effects of CP-101,606 were associated with a reversal of the CUMS-induced decrease in mTOR (Ser2448) and p70S6K (Thr389) phosphorylation and increasing PSD95 and GluA1 synthesis in the hippocampus. CONCLUSIONS Our results demonstrate that CP-101,606 produces antidepressant effects in CUMS mice, which may be mediated by mTOR signaling cascade upregulation. Our findings suggest the possible utility of CP-101,606 as a treatment for depression.
Collapse
Affiliation(s)
- Yu Qin
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xinlei Guo
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Wenyue Song
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zehuai Liang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yahui Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Dan Feng
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yiru Yang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Mingxing Li
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Mingqi Gao
- School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
48
|
Vila-Merkle H, González-Martínez A, Campos-Jiménez R, Martínez-Ricós J, Teruel-Martí V, Lloret A, Blasco-Serra A, Cervera-Ferri A. Sex differences in amygdalohippocampal oscillations and neuronal activation in a rodent anxiety model and in response to infralimbic deep brain stimulation. Front Behav Neurosci 2023; 17:1122163. [PMID: 36910127 PMCID: PMC9995972 DOI: 10.3389/fnbeh.2023.1122163] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Depression and anxiety are highly comorbid mental disorders with marked sex differences. Both disorders show altered activity in the amygdala, hippocampus, and prefrontal cortex. Infralimbic deep brain stimulation (DBS-IL) has anxiolytic and antidepressant effects, but the underlying mechanisms remain unclear. We aimed to contribute to understanding sex differences in the neurobiology of these disorders. Methods In male and female rats, we recorded neural oscillations along the dorsoventral axis of the hippocampus and the amygdala in response to an anxiogenic drug, FG-7142. Following this, we applied DBS-IL. Results Surprisingly, in females, the anxiogenic drug failed to induce most of the changes observed in males. We found sex differences in slow, delta, theta, and beta oscillations, and the amygdalo-hippocampal communication in response to FG-7142, with modest changes in females. Females had a more prominent basal gamma, and the drug altered this band only in males. We also analyzed c-Fos expression in both sexes in stress-related structures in response to FG-7142, DBS-IL, and combined interventions. With the anxiogenic drug, females showed reduced expression in the nucleus incertus, amygdala, septohippocampal network, and neocortical levels. In both experiments, the DBS-IL reversed FG-7142-induced effects, with a more substantial effect in males than females. Discussion Here, we show a reduced response in female rats which contrasts with the higher prevalence of anxiety in women but is consistent with other studies in rodents. Our results open compelling questions about sex differences in the neurobiology of anxiety and depression and their study in animal models.
Collapse
Affiliation(s)
- Hanna Vila-Merkle
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Alicia González-Martínez
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Rut Campos-Jiménez
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Joana Martínez-Ricós
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Vicent Teruel-Martí
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Ana Lloret
- Department of Physiology, Faculty of Medicine, Health Research Institute INCLIVA, CIBERFES, University of Valencia, Valencia, Spain
| | - Arantxa Blasco-Serra
- Study Group for the Anatomical Substrate of Pain and Analgesia (GESADA) Laboratory, Department of Human Anatomy and Embryology, Faculty of Medicine and Odontology, University of Valencia, Valencia, Spain
| | - Ana Cervera-Ferri
- Neuronal Circuits Laboratory, Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
49
|
Meshkat S, Ho RC, Cao B, Teopiz KM, Rosenblat JD, Rhee TG, Di Vincenzo JD, Ceban F, Jawad MY, McIntyre RS. Biomarkers of ketamine's antidepressant effect: An umbrella review. J Affect Disord 2023; 323:598-606. [PMID: 36521662 DOI: 10.1016/j.jad.2022.12.021] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Abstract
Ketamine is a NMDA receptor antagonist that has a rapid acting antidepressant effect with high efficacy in treatment-resistant patients. Ketamine is a beneficial antidepressant for many individuals with depression, but not all of the patients respond, and some even exhibit symptom deterioration. The discovery of repeatable and mechanistically relevant biomarkers would address a major gap in treatment response prediction. Numerous potential peripheral biomarkers have been reported, but their current utility is unclear. We conducted an umbrella review to evaluate the biomarkers of ketamine's antidepressant effect in individuals with depression. PubMed and copus were searched using terms appropriate to each area of research, from their inception until July 2022. Five systematic reviews and meta analyses including 108 studies with 4912 participants were included. Blood-based and neuroimaging biomarkers were investigated. The results of this review indicate that ketamine can produce an anti-inflammatory effect and decrease at least one inflammatory marker following administration. Data from neuroimaging studies demonstrated that the cingulate cortex is the key locus of ketamine's action. The majority of the blood-based, neuroimaging, and neurophysiological investigations reviewed herein indicate ketamine induced normalization of major depressive disorder pathogenesis via synaptic plasticity and functional connectivity. Currently, no biomarker/biosignature is sufficiently validated for clinical utility, but several are promising. Now that ketamine is more widely available, biomarker discovery and replication should be attempted in larger, real-world populations.
Collapse
Affiliation(s)
- Shakila Meshkat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Roger C Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, PR China
| | - Kayla M Teopiz
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Taeho Greg Rhee
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA; Department of Public Health Sciences, School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Felicia Ceban
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Muhammad Youshay Jawad
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| |
Collapse
|
50
|
Rossi GN, Hallak JEC, Baker G, Dursun SM, Dos Santos RG. The effects of ketamine and classic hallucinogens on neurotrophic and inflammatory markers in unipolar treatment-resistant depression: a systematic review of clinical trials. Eur Arch Psychiatry Clin Neurosci 2023; 273:129-155. [PMID: 35829812 DOI: 10.1007/s00406-022-01460-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/27/2022] [Indexed: 11/03/2022]
Abstract
Although results are still preliminary, ketamine and classical hallucinogens have shown promise in recent years as novel, fast-acting antidepressants, especially for the treatment of unipolar treatment-resistant depression (TRD). Depression also seems to be related to abnormal levels of peripheral inflammatory and neurotrophic biomarkers, which may one day help to diagnose of this disorder. In this context, this systematic review of clinical trials evaluated the current evidence that relates the antidepressant effects of ketamine and classical hallucinogens on TRD with changes in inflammatory and neurotrophic biomarkers. Twelve studies were found (n = 587), 2 with oral ayahuasca (1 mL/kg) and 10 with ketamine (mostly intravenous 0.5 mg/kg) administration. Results for all biomarkers assessed were contradictory and thus inconclusive. Randomized controlled trials with bigger samples and higher statistical power are warranted to clarify if peripheral biomarkers can confidently be used to indicate and measure ketamine's and classical hallucinogens' antidepressant effect. The PROSPERO ID for this study is CRD42021249089.
Collapse
Affiliation(s)
- Giordano Novak Rossi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Jaime E C Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen Baker
- National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Serdar M Dursun
- National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Rafael G Dos Santos
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil. .,National Institute for Translational Medicine (INCT-TM), CNPq, São Paulo, Brazil. .,Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Hospital das Clínicas, Terceiro Andar, Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|